1
|
Bogdan C, Islam NAK, Barinberg D, Soulat D, Schleicher U, Rai B. The immunomicrotope of Leishmania control and persistence. Trends Parasitol 2024; 40:788-804. [PMID: 39174373 DOI: 10.1016/j.pt.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024]
Abstract
Leishmania is an intracellular protozoan transmitted by sand fly vectors; it causes cutaneous, mucocutaneous, or visceral disease. Its growth and survival are impeded by type 1 T helper cell responses, which entail interferon (IFN)-γ-mediated macrophage activation. Leishmania partially escapes this host defense by triggering immune cell and cytokine responses that favor parasite replication rather than killing. Novel methods for in situ analyses have revealed that the pathways of immune control and microbial evasion are strongly influenced by the tissue context, the micro milieu factors, and the metabolism at the site of infection, which we collectively term the 'immunomicrotope'. Understanding the components and the impact of the immunomicrotope will enable the development of novel strategies for the treatment of chronic leishmaniasis.
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany.
| | - Noor-A-Kasida Islam
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - David Barinberg
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Baplu Rai
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| |
Collapse
|
2
|
Evans C, Mutasa K, Rukobo S, Govha M, Mushayanembwa P, Chasekwa B, Majo FD, Tavengwa NV, Broad J, Noble C, Gough EK, Kelly P, Bourke CD, Humphrey JH, Ntozini R, Prendergast AJ. Inflammation and cytomegalovirus viremia during pregnancy drive sex-differentiated differences in mortality and immune development in HIV-exposed infants. Nat Commun 2024; 15:2909. [PMID: 38632279 PMCID: PMC11024190 DOI: 10.1038/s41467-023-44166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 12/04/2023] [Indexed: 04/19/2024] Open
Abstract
Children who are HIV-exposed but uninfected have increased infectious mortality compared to HIV-unexposed children, raising the possibility of immune abnormalities following exposure to maternal viraemia, immune dysfunction, and co-infections during pregnancy. In a secondary analysis of the SHINE trial in rural Zimbabwe we explored biological pathways underlying infant mortality, and maternal factors shaping immune development in HIV-exposed uninfected infants. Maternal inflammation and cytomegalovirus viraemia were independently associated with infant deaths: mortality doubled for each log10 rise in maternal C-reactive protein (adjusted hazard ratio (aHR) 2.09; 95% CI 1.33-3.27), and increased 1.6-fold for each log10 rise in maternal cytomegalovirus viral load (aHR 1.62; 95% CI 1.11-2.36). In girls, mortality was more strongly associated with maternal C-reactive protein than cytomegalovirus; in boys, mortality was more strongly associated with cytomegalovirus than C-reactive protein. At age one month, HIV-exposed uninfected infants had a distinct immune milieu, characterised by raised soluble CD14 and an altered CD8 + T-cell compartment. Alterations in immunophenotype and systemic inflammation were generally greater in boys than girls. Collectively, these findings show how the pregnancy immune environment in women with HIV underlies mortality and immune development in their offspring in a sex-differentiated manner, and highlights potential new intervention strategies to transform outcomes of HIV-exposed children. ClinicalTrials.gov/NCT01824940.
Collapse
Affiliation(s)
- Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe.
- Blizard Institute, Queen Mary University of London, London, UK.
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Sandra Rukobo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Margaret Govha
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jonathan Broad
- Blizard Institute, Queen Mary University of London, London, UK
| | - Christie Noble
- Blizard Institute, Queen Mary University of London, London, UK
| | - Ethan K Gough
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Paul Kelly
- Blizard Institute, Queen Mary University of London, London, UK
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Claire D Bourke
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Blizard Institute, Queen Mary University of London, London, UK
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Blizard Institute, Queen Mary University of London, London, UK
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
3
|
Watany MM, Elhosary MM. Clinical utility of circulating TWEAK and CD163 as biomarkers of iron-induced cardiac decompensation in transfusion dependent thalassemia major. Cytokine 2024; 173:156443. [PMID: 38000169 DOI: 10.1016/j.cyto.2023.156443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/30/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND AND AIM Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) affects most of the cells involved in cardiac fibrosis like inflammatory cells, cardiomyocytes and fibroblasts. CD163, the receptor of TWEAK on the surface of type 2 macrophages, is shed into plasma upon macrophages activation. This work aimed to evaluate serum TWEAK and its decoy receptor CD163 as probable biomarkers to monitor myocardial iron overload (MIO) in transfusion dependent thalassemia major (TDTM) patients and to predict iron-induced cardiac decompensation (IICD). METHODS A total of 140 TDTM patients were enrolled. Patients were categorized into two groups; group I (n = 70) diagnosed with IICD while group II (n = 70) had no evidence of IICD. sTWEAK and sCD163 were quantitated utilizing Enzyme-linked-immunosorbent- assay. RESULTS sTWEAK was evidently lower in group I than group II (medians, 412 and 1052 pg/mL respectively). sCD163 was higher in group I than group II (medians, 615.5 and 323.5 ng/mL respectively). sTWEAK positively correlated with cardiac MRI-T2 mapping and ventricular ejection fractions and negatively correlated with B-Natriuretic peptide and cardiac troponin. An inverse relationship between TWEAK and CD163 was documented throughout the study. sTWEAK, sCD163 and TWEAK/CD163 ratio proved to be significant predictors of IICD in TDTM patients. TWEAK/CD163 ratio < 1.04 discriminated IICD in TDTM patients with 100 % clinical sensitivity and specificity. CONCLUSION Circulating TWEAK and CD163 appears to be promising biomarkers for monitoring MIO and predicting IICD in TDTM patients.
Collapse
Affiliation(s)
- Mona M Watany
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Marwa M Elhosary
- Msc Immunology from Tanta University, Faculty of Science, Tanta 31527, Egypt
| |
Collapse
|
4
|
Cao Y, Liang N, Kong K, Qiao X, Liu T, Fang JA, Zhang X. CD163 as a Potential Biomarker-associated Immune Inflammation in Diabetes Mellitus: A Systematic Review and Bioinformatics Analysis. Endocr Metab Immune Disord Drug Targets 2024; 24:208-219. [PMID: 37455460 DOI: 10.2174/1871530323666230714162324] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/20/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Several studies have identified CD163 as a potential mediator of diabetes mellitus through an immune-inflammation. Further study is necessary to identify its specific mechanism. OBJECTIVES In this study, we aimed to investigate CD163 as a potential biomarker associated with immune inflammation in diabetes mellitus through a systematic review and bioinformatics analysis. METHODS We searched PubMed, Web of Science, the Cochrane Library, and Embase databases with a time limit of September 2, 2022. Furthermore, we conducted a systematic search and review based on PRISMA guidelines. Additionally, diabetic gene expression microarray datasets GSE29221, GSE30528, GSE30529, and GSE20966 were downloaded from the GEO database (http://www.ncbi.nlm.nih.gov/geo) for bioinformatics analysis. The PROSPERO number for this study is CRD420222347160. RESULTS Following the inclusion and exclusion criteria, seven articles included 1607 patients, comprising 912 diabetic patients and 695 non-diabetic patients. This systematic review found significantly higher levels of CD163 in diabetic patients compared to non-diabetic patients. People with diabetes had higher levels of CRP expression compared to the control group. Similarly, two of the three papers that used TNF- α as an outcome indicator showed higher expression levels in diabetic patients. Furthermore, IL-6 expression levels were higher in diabetic patients than in the control group. A total of 62 samples were analyzed by bioinformatics (33 case controls and 29 experimental groups), and 85 differential genes were identified containing CD163. According to the immune cell correlation analysis, CD163 was associated with macrophage M2, γδ T lymphocytes, macrophage M1, and other immune cells. Furthermore, to evaluate the diagnostic performance of CD163, we validated it using the GSE20966 dataset. In the validation set, CD163 showed high diagnostic accuracy. CONCLUSION This study suggests CD163 participates in the inflammatory immune response associated with diabetes mellitus and its complications by involving several immune cells. Furthermore, the results suggest CD163 may be a potential biomarker reflecting immune inflammation in diabetic mellitus.
Collapse
Affiliation(s)
- Yang Cao
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Ning Liang
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Kaili Kong
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiaomei Qiao
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Ting Liu
- Department of Nephrology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing-Ai Fang
- Department of Nephrology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaodong Zhang
- Department of Nephrology, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
5
|
Ravi S, Duraisamy P, Krishnan M, Martin LC, Manikandan B, Ramar M. Sitosterol-rich Digera muricata against 7-ketocholesterol and lipopolysaccharide-mediated atherogenic responses by modulating NF-ΚB/iNOS signalling pathway in macrophages. 3 Biotech 2023; 13:331. [PMID: 37670802 PMCID: PMC10475456 DOI: 10.1007/s13205-023-03741-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023] Open
Abstract
Digera muricata L., commonly known as Tartara, is an edible herb used as traditional medicine in many countries of Africa and Asia. This study aimed to elucidate the effect of a phytosterol-rich extract of D. muricata on 7-ketocholesterol-mediated atherosclerosis in macrophages. The extract was examined by phytochemical analyses, GC-MS, TLC, DPPH scavenging and hRBC membrane stabilization assays. Macrophage polarization was studied with experimental groups framed based on alamar blue cell viability and griess assays. Regulations of arginase enzyme activity, ROS generation, mitochondrial membrane potential, cell membrane integrity, pinocytosis, lipid uptake and peroxidation, as well as, intracellular calcium deposition were determined. In addition, expressions of atherogenic mediators were analysed using PCR, ELISA and immunocytochemistry techniques. Diverse phytochemicals with higher free radical scavenging activity and anti-inflammatory potential have been detected in the D. muricata. Co-treatment with D. muricata markedly reduced the atherogenic responses induced by 7KCh in the presence of LPS such as ROS, especially, NO and O2- along with lipid peroxidation. Furthermore, D. muricata significantly normalized mitochondrial membrane potential, cell membrane integrity, pinocytic activity, intracellular lipid accumulation and calcium deposition. These results provided us with the potentiality of D. muricata in ameliorating atherogenesis. Additionally, it decreased the expression of pro-atherogenic mediators (iNOS, COX-2, MMP9, IL-6, IL-1β, CD36, CD163 and TGFβ1) and increased anti-atherogenic mediators (MRC1 and PPARγ) with high cellular expressions of NF-κB and iNOS. Results showed the potential of sitosterol-rich D. muricata as a versatile biomedical therapeutic agent against abnormal macrophage polarization and its associated pathologies.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni’s College for Women, Chennai, 600 015 India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| |
Collapse
|
6
|
Rakić M, Lunić T, Bekić M, Tomić S, Mitić K, Graovac S, Božić B, Božić Nedeljković B. Vitamin B complex suppresses neuroinflammation in activated microglia: in vitro and in silico approach combined with dynamical modeling. Int Immunopharmacol 2023; 121:110525. [PMID: 37356121 DOI: 10.1016/j.intimp.2023.110525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/22/2023] [Accepted: 06/14/2023] [Indexed: 06/27/2023]
Abstract
Activated microglia is critically involved in the regulation of neuroinflammation/neurodegradation. Hereby, the anti-inflammatory effects of the vitamin B complex (VBC - B1, B2, B3, B5, B6, and B12) on the function and phenotype of lipopolysaccharide (LPS)-stimulated BV2 microglial cells were examined in vitro. Additionally, VBC-treated microglia supernatants were evaluated on SH-SY5Y cells to investigate the effects on neurons' viability. Further, anti-inflammatory mechanisms of VBC were examined by molecular dockingstudies to determine the binding affinity of each VBC component to Toll-like receptor 4 (TLR4) signalling pathway proteins and inducible nitric oxide synthase. In addition, the dynamical model which simulates VBC inhibition of TLR4 signalling pathway proteins activated by LPS has been constructed and excellent agreement with experimental data has been observed (adjR2 = 0.9715 and 0.9909 for TNF-α and IL-6, respectively). The obtained data demonstrated that VBC treatment reduced the inflammatory mediators secreted by LPS-stimulated microglia, diminished their neurotoxic effects against neurons, and induced changes in phenotype profile toward M2 microglia type. Finally, the constructed dynamical model provides deeper insight into the involvement of each VBC component on the VBC inhibitory potential toward the TLR4 signalling pathway and enables optimization of novel VBC formulations as well as inhibitory potential of new putative inhibitors.
Collapse
Affiliation(s)
- Marija Rakić
- University of Belgrade, Faculty of Biology, 11000 Belgrade, Serbia.
| | - Tanja Lunić
- University of Belgrade, Faculty of Biology, 11000 Belgrade, Serbia.
| | - Marina Bekić
- University of Belgrade, Institute for the Application of Nuclear Energy, INEP, 11080 Belgrade, Serbia.
| | - Sergej Tomić
- University of Belgrade, Institute for the Application of Nuclear Energy, INEP, 11080 Belgrade, Serbia.
| | - Katarina Mitić
- University of Belgrade, Faculty of Biology, 11000 Belgrade, Serbia.
| | - Stefan Graovac
- University of Belgrade, Faculty of Physics, 11000 Belgrade, Serbia.
| | - Bojan Božić
- University of Belgrade, Faculty of Biology, 11000 Belgrade, Serbia.
| | | |
Collapse
|
7
|
Perkins MV, Joseph S, Dittmer DP, Mackman N. Cardiovascular Disease and Thrombosis in HIV Infection. Arterioscler Thromb Vasc Biol 2023; 43:175-191. [PMID: 36453273 PMCID: PMC10165851 DOI: 10.1161/atvbaha.122.318232] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
HIV infection has transitioned from an acute, fatal disease to a chronic one managed by antiretroviral therapy. Thus, the aging population of people living with HIV (PLWH) continues to expand. HIV infection results in a dysregulated immune system, wherein CD4+ T cells are depleted, particularly in the gastrointestinal tract, disrupting the gut epithelial barrier. Long-term HIV infection is associated with chronic inflammation through potentially direct mechanisms caused by viral replication or exposure to viral proteins and indirect mechanisms resulting from increased translocation of microbial products from the intestine or exposure to antiretroviral therapy. Chronic inflammation (as marked by IL [interleukin]-6 and CRP [C-reactive protein]) in PLWH promotes endothelial cell dysfunction and atherosclerosis. PLWH show significantly increased rates of cardiovascular disease, such as myocardial infarction (risk ratio, 1.79 [95% CI, 1.54-2.08]) and stroke (risk ratio, 2.56 [95% CI, 1.43-4.61]). In addition, PLWH have increased levels of the coagulation biomarker D-dimer and have a two to ten-fold increased risk of venous thromboembolism compared with the general population. Several small clinical trials analyzed the effect of different antithrombotic agents on platelet activation, coagulation, inflammation, and immune cell activation. Although some markers for coagulation were reduced, most agents failed to reduce inflammatory markers in PLWH. More studies are needed to understand the underlying mechanisms driving inflammation in PLWH to create better therapies for lowering chronic inflammation in PLWH. Such therapies can potentially reduce atherosclerosis, cardiovascular disease, and thrombosis rates in PLWH and thus overall mortality in this population.
Collapse
Affiliation(s)
- Megan V. Perkins
- UNC Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah Joseph
- UNC Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dirk P. Dittmer
- UNC Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Stuhr LK, Madsen K, Johansen AZ, Chen IM, Hansen CP, Jensen LH, Hansen TF, Kløve-Mogensen K, Nielsen KR, Johansen JS. Combining sCD163 with CA 19-9 Increases the Predictiveness of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:cancers15030897. [PMID: 36765852 PMCID: PMC9913074 DOI: 10.3390/cancers15030897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/22/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
The objective of this study was to evaluate the diagnostic and prognostic potential of soluble CD163 (sCD163) in patients with pancreatic ductal adenocarcinoma (PDAC). Preoperative serum samples from 255 patients with PDAC were analyzed for sCD163 using a commercially available enzyme-linked immunosorbent assay. The diagnostic value of sCD163 was evaluated using receiver operating characteristic (ROC) curves. The prognostic significance of sCD163 was evaluated by Cox regression analysis and Kaplan-Meier survival curves. sCD163 was significantly increased in patients with PDAC, across all stages, compared to healthy subjects (stage 1: p value = 0.033; stage 2-4: p value ≤ 0.0001). ROC curves showed that sCD163 combined with CA 19-9 had the highest diagnostic potential compared to sCD163 and CA 19-9 alone both in patients with local PDAC and patients with advanced PDAC. Univariate and multivariate analysis showed no association between sCD163 and overall survival. This study found elevated levels of circulating sCD163 in patients with PDAC, regardless of stage, compared to healthy subjects. This suggests that sCD163 may have a clinical value as a novel diagnostic biomarker in PDAC.
Collapse
Affiliation(s)
- Liva K. Stuhr
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, DK-2730 Herlev, Denmark
| | - Kasper Madsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, DK-2730 Herlev, Denmark
| | - Astrid Z. Johansen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, DK-2730 Herlev, Denmark
| | - Inna M. Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, DK-2730 Herlev, Denmark
| | - Carsten P. Hansen
- Department of Surgery, Copenhagen University Hospital-Rigshospitalet, DK-2200 Copenhagen, Denmark
| | - Lars H. Jensen
- Department of Oncology, University Hospital of Southern Denmark, DK-7100 Vejle, Denmark
| | - Torben F. Hansen
- Department of Oncology, University Hospital of Southern Denmark, DK-7100 Vejle, Denmark
| | | | - Kaspar R. Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | - Julia S. Johansen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, DK-2730 Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital-Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Correspondence: ; Tel.: +45-38689241
| |
Collapse
|
9
|
Mierzchała-Pasierb M, Lipińska-Gediga M, Lewandowski Ł, Krzystek-Korpacka M. Alterations in Serum Concentration of Soluble CD163 within Five Study Days from ICU Admission Are Associated with In-Hospital Mortality of Septic Patients-A Preliminary Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2263. [PMID: 36767629 PMCID: PMC9916291 DOI: 10.3390/ijerph20032263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND CD163, a cell membrane surface molecule specifically expressed by macrophages with an anti-inflammatory phenotype, participates in innate immunity. The purpose of the study was to evaluate the clinical utility of sCD163 in septic patients in comparison to other parameters associated with infections, mainly PCT, CRP and IL-18. METHODS Serum samples were obtained from 40 septic patients on the ICU admission day, 3rd and 5th study days. The control group consisted of 30 healthy volunteers from whom the specimen was collected once. An enzyme-linked immunosorbent assay (ELISA) was used to determine the concentrations of sCD163 and IL-18. CRP and PCT records, among others, were provided by the hospital. RESULTS Septic shock was associated with the highest concentrations of sCD163 and IL-18. Admission values of sCD163 significantly contributed to mortality prediction in septic patients. CONCLUSIONS The concentration of sCD163 determined on the ICU admission day may potentially be utilized in estimation of the odds of death among septic patients.
Collapse
Affiliation(s)
| | | | - Łukasz Lewandowski
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | | |
Collapse
|
10
|
Chitin Derived Small Molecule AVR-48 Reprograms the Resting Macrophages to an Intermediate Phenotype and Decrease Pseudomonas aeruginosa Mouse Lung Infection. IMMUNO 2022. [DOI: 10.3390/immuno2040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
AVR-48 is a structural derivative of chitin previously shown by our laboratory to significantly decrease lung injury parameters in LPS, hyperoxia and sepsis-induced rodent models. The current study objectives are to determine the cellular mechanism of action and demonstrate efficacy in a mouse bacterial lung infection model. For in vitro receptor binding and macrophage polarization studies, C57Bl/6J mouse derived spleens and human peripheral blood mononuclear cells (hPBMCs) were treated with AVR-48 ± LPS or biotin conjugated AVR-48. Different macrophage types were determined using flow cytometry and secreted cytokines were measured using ELISA. In vivo, a CD-1 mouse Pseudomonas aeruginosa lung infection was treated with AVR-48, assessing bacterial colony forming unit (CFU), IL-10 and IL-17A levels in lung and blood samples. AVR-48 binds to both the toll-like receptor 4 (TLR4) and the CD163 receptor on mouse monocytes. In hPBMCs, frequency of intermediate macrophages increased upon AVR-48 treatment for 72 h. Increased bacterial phagocytosis/intracellular killing were observed in THP-1 cells and reduction in CFU in CD-1 mouse lungs. Binding of AVR-48 to both TLR4 and CD163 receptors bring the macrophages to an intermediary stage, resulting in increased phagocytosis and decreased inflammation, altogether providing an optimal immune balance for treating lung injury and infection.
Collapse
|
11
|
Peña AN, Sommerfeld SD, Anderson AE, Han J, Maestas DR, Mejias JC, Woodell-May J, King W, Ganguly S, Elisseeff JH. Autologous Protein Solution processing alters lymphoid and myeloid cell populations and modulates gene expression dependent on cell type. Arthritis Res Ther 2022; 24:221. [PMID: 36096945 PMCID: PMC9465964 DOI: 10.1186/s13075-022-02875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/21/2022] [Indexed: 11/10/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease associated with cartilage degradation, osteophyte formation, and fibrillation. Autologous Protein Solution (APS), a type of autologous anti-inflammatory orthobiologic, is used for pain management and treatment of OA. Various compositions of autologous PRP formulations are in clinical use for musculoskeletal pathologies, by nature of their minimal processing and source of bioactive molecules. Currently, there is no consensus on the optimal composition of the complex mixture. In this study, we focused on elucidating the immune cell subtypes and phenotypes in APS. We identified the immune cell types in APS from healthy donors and investigated phenotypic changes in the immune cells after APS processing. Based on flow cytometric analysis, we found that neutrophils and T cells are the most abundant immune cell types in APS, while monocytes experience the largest fold change in concentration compared to WBCs. Gene expression profiling revealed that APS processing results in differential gene expression changes dependent on immune cell type, with the most significantly differentially regulated genes occurring in the monocytes. Our results demonstrate that the mechanical processing of blood, whose main purpose is enrichment and separation, can alter its protein and cellular composition, as well as cellular phenotypes in the final product.
Collapse
Affiliation(s)
- Alexis N Peña
- Translational Tissue Engineering Center, Johns Hopkins University, 400 N. Broadway Smith Building 5th floor, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Sven D Sommerfeld
- Translational Tissue Engineering Center, Johns Hopkins University, 400 N. Broadway Smith Building 5th floor, Baltimore, MD, 21231, USA
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Amy E Anderson
- Translational Tissue Engineering Center, Johns Hopkins University, 400 N. Broadway Smith Building 5th floor, Baltimore, MD, 21231, USA
- Department of Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jin Han
- Translational Tissue Engineering Center, Johns Hopkins University, 400 N. Broadway Smith Building 5th floor, Baltimore, MD, 21231, USA
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - David R Maestas
- Translational Tissue Engineering Center, Johns Hopkins University, 400 N. Broadway Smith Building 5th floor, Baltimore, MD, 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Joscelyn C Mejias
- Translational Tissue Engineering Center, Johns Hopkins University, 400 N. Broadway Smith Building 5th floor, Baltimore, MD, 21231, USA
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | | | - William King
- Zimmer Biomet, 56 East Bell Drive, Warsaw, IN, 46581, USA
| | - Sudipto Ganguly
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Johns Hopkins University, 400 N. Broadway Smith Building 5th floor, Baltimore, MD, 21231, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
12
|
Kazmierski J, Friedmann K, Postmus D, Emanuel J, Fischer C, Jansen J, Richter A, Bosquillon de Jarcy L, Schüler C, Sohn M, Sauer S, Drosten C, Saliba A, Sander LE, Müller MA, Niemeyer D, Goffinet C. Nonproductive exposure of PBMCs to SARS-CoV-2 induces cell-intrinsic innate immune responses. Mol Syst Biol 2022; 18:e10961. [PMID: 35975552 PMCID: PMC9382356 DOI: 10.15252/msb.202210961] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Cell-intrinsic responses mounted in PBMCs during mild and severe COVID-19 differ quantitatively and qualitatively. Whether they are triggered by signals emitted by productively infected cells of the respiratory tract or result from physical interaction with virus particles remains unclear. Here, we analyzed susceptibility and expression profiles of PBMCs from healthy donors upon ex vivo exposure to SARS-CoV and SARS-CoV-2. In line with the absence of detectable ACE2 receptor expression, human PBMCs were refractory to productive infection. RT-PCR experiments and single-cell RNA sequencing revealed JAK/STAT-dependent induction of interferon-stimulated genes (ISGs) but not proinflammatory cytokines. This SARS-CoV-2-specific response was most pronounced in monocytes. SARS-CoV-2-RNA-positive monocytes displayed a lower ISG signature as compared to bystander cells of the identical culture. This suggests a preferential invasion of cells with a low ISG baseline profile or delivery of a SARS-CoV-2-specific sensing antagonist upon efficient particle internalization. Together, nonproductive physical interaction of PBMCs with SARS-CoV-2- and, to a much lesser extent, SARS-CoV particles stimulate JAK/STAT-dependent, monocyte-accentuated innate immune responses that resemble those detected in vivo in patients with mild COVID-19.
Collapse
Affiliation(s)
- Julia Kazmierski
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Kirstin Friedmann
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Dylan Postmus
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Jackson Emanuel
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Cornelius Fischer
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Jenny Jansen
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Anja Richter
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Laure Bosquillon de Jarcy
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Department of Infectious Diseases and Respiratory MedicineCharité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health (BIH)BerlinGermany
| | - Christiane Schüler
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Madlen Sohn
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Sascha Sauer
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Christian Drosten
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Antoine‐Emmanuel Saliba
- Helmholtz Institute for RNA‐based Infection Research (HIRI)Helmholtz‐Center for Infection Research (HZI)WürzburgGermany
| | - Leif Erik Sander
- Department of Infectious Diseases and Respiratory MedicineCharité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health (BIH)BerlinGermany
| | - Marcel A Müller
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Daniela Niemeyer
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Christine Goffinet
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| |
Collapse
|
13
|
Chauvin P, Morzadec C, de Latour B, Llamas-Gutierrez F, Luque-Paz D, Jouneau S, Vernhet L. Soluble CD163 is produced by monocyte-derived and alveolar macrophages, and is not associated with the severity of idiopathic pulmonary fibrosis. Innate Immun 2022; 28:138-151. [PMID: 35522300 PMCID: PMC9136464 DOI: 10.1177/17534259221097835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022] Open
Abstract
The soluble form of the membrane hemoglobin scavenger receptor CD163 (sCD163), released by shedding, is a strong marker for macrophage activation. Serum sCD163 levels rise in several acute inflammatory states and some fibrosing diseases. Monocyte-derived macrophages (MoDM) differentiated by macrophage colony-stimulating factor (M-MoDM) contribute to the pathophysiology of idiopathic pulmonary fibrosis (IPF), an irreversible and rapidly fatal interstitial lung disease. Since M-MoDM express high membrane CD163 levels, we thus postulated that sCD163 could be a relevant biomarker for macrophage activation in IPF. We found that M-MoDM constitutively released higher amounts of sCD163 (49.5 ± 24.5 ng/ml) than monocytes (0.45 ± 0.32 ng/ml) or MoDM differentiated with granulocyte macrophage-stimulating factor (2.24 ± 0.98 ng/ml). The basal production of sCD163 by M-MoDM was increased following stimulation with lipopolysaccharide (123.4 ± 54.9 ng/ml) or ATP (168.9 ± 41.8 ng/ml). The sCD163 release was controlled by metalloproteases but not through ADAM17 activation. Moreover, CD163-positive macrophages and sCD163 were detected in pulmonary tissues and alveolar fluids of Caucasian patients with IPF, respectively. IPF alveolar macrophages constitutively secreted sCD163 amounts (67.6 ± 44.6 ng/µg RNA) which were significantly higher than those released by alveolar macrophages isolated from controls (19.2 ± 7.6 ng/µg RNA) or patients with other interstitial lung disease (31.5 ± 16.6 ng/µg RNA). However, the concentrations of sCD163 in blood serum collected from 155 patients with IPF did not correlate with the severity of their disease. In conclusion, our results show that M-MoDM constituted a pertinent model to study the regulation of sCD163 production. Yet, serum sCD163 values could not provide a prognostic biomarker for IPF in our cohort.
Collapse
Affiliation(s)
- Pierre Chauvin
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche
en santé, environnement et travail), Rennes, France
| | - Claudie Morzadec
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé,
environnement et travail), Rennes, France
| | - Bertrand de Latour
- Service de Chirurgie cardio-thoracique et vasculaire, Centre Hospitalier Universitaire,
Rennes, France
| | | | - David Luque-Paz
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche
en santé, environnement et travail), Rennes, France
| | - Stéphane Jouneau
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche
en santé, environnement et travail), Rennes, France
- Service de Pneumologie, Centre de compétences pour les maladies
pulmonaires rares de Bretagne, Centre Hospitalier Universitaire,
Rennes, France
| | - Laurent Vernhet
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé,
environnement et travail), Rennes, France
| |
Collapse
|
14
|
Underwood ML, Park B, Uebelhoer LS, Gu G, Kunkel LE, Korthuis PT, Cook RR, Sekaly RP, Ribeiro SP, Lancioni CL. Chronic Alcohol Exposure Among People Living with HIV Is Associated with Innate Immune Activation and Alterations in Monocyte Phenotype and Plasma Cytokine Profile. Front Immunol 2022; 13:867937. [PMID: 35371104 PMCID: PMC8971672 DOI: 10.3389/fimmu.2022.867937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/23/2022] [Indexed: 12/25/2022] Open
Abstract
Despite advances in antiretroviral therapy, chronic immune activation continues to be observed among individuals with well-controlled HIV viral loads, and is associated with non-AIDS defining morbidities among people living with HIV. Alcohol use disorder impacts a significant proportion of individuals living with HIV, and alcohol exposure is known to damage the intestinal epithelium which may increase translocation of pathogens and their molecular products, driving systemic immune activation and dysregulation. The aim of this study was to determine if adults living with HIV with well-controlled viral loads, who also suffer from alcohol use disorder with and without hepatitis C virus co-infection (n=23), exhibit evidence of advanced systemic immune activation, intestinal damage, and microbial translocation, as compared to adults living with HIV who are not exposed to chronic alcohol or other substances of abuse (n=29). The impact of a 1-month intervention to treat alcohol-use disorder was also examined. Alcohol-use disorder was associated with evidence of advanced innate immune activation, alterations in monocyte phenotype including increased expression of Toll-like receptor 4, increased burden of stimulatory ligands for Toll-like receptor 4, and alterations in plasma cytokine signature, most notably elevations in soluble CD40 ligand and transforming growth factor beta. Alcohol-associated immune activation was more pronounced among individuals with hepatitis C virus co-infection. Although the 1-month intervention to treat alcohol use disorder did not result in significant reductions in the interrogated indicators of immune activation, our findings suggest that chronic alcohol exposure is a major modifiable risk factor for chronic immune activation and dysregulation among people-living with HIV.
Collapse
Affiliation(s)
- Michelle L. Underwood
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
| | - Byung Park
- Knight-Cancer Institute, Department of Biostatistics, Oregon Health & Science University, Portland, OR, United States
| | - Luke S. Uebelhoer
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
| | - Geoffrey Gu
- Undergraduate Studies, University of Southern California, Los Angeles, CA, United States
| | - Lynn E. Kunkel
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Philip T. Korthuis
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
- Department of Public Health, Oregon Health & Science University, Portland, OR, United States
| | - Ryan R. Cook
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Rafick Pierre Sekaly
- Department of Pathology & Translational Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Susan Pereira Ribeiro
- Department of Pathology & Translational Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Christina L. Lancioni
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
- *Correspondence: Christina L. Lancioni,
| |
Collapse
|
15
|
Potor L, Hendrik Z, Patsalos A, Katona É, Méhes G, Póliska S, Csősz É, Kalló G, Komáromi I, Combi Z, Posta N, Sikura KÉ, Pethő D, Oros M, Vereb G, Tóth C, Gergely P, Nagy L, Balla G, Balla J. Oxidation of Hemoglobin Drives a Proatherogenic Polarization of Macrophages in Human Atherosclerosis. Antioxid Redox Signal 2021; 35:917-950. [PMID: 34269613 PMCID: PMC8905252 DOI: 10.1089/ars.2020.8234] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Aim: The aim of our study was to explore the pathophysiologic role of oxidation of hemoglobin (Hb) to ferrylHb in human atherosclerosis. Results: We observed a severe oxidation of Hb to ferrylHb in complicated atherosclerotic lesions of carotid arteries with oxidative changes of the globin moieties, detected previously described oxidation hotspots in Hb (β1Cys93; β1Cys112; β2Cys112) and identified a novel oxidation hotspot (α1Cys104). After producing a monoclonal anti-ferrylHb antibody, ferrylHb was revealed to be localized extracellularly and also internalized by macrophages in the human hemorrhagic complicated lesions. We demonstrated that ferrylHb is taken up via phagocytosis as well as CD163 receptor-mediated endocytosis and then transported to lysosomes involving actin polymerization. Internalization of ferrylHb was accompanied by upregulation of heme oxygenase-1 and H-ferritin and accumulation of iron within lysosomes as a result of heme/iron uptake. Importantly, macrophages exposed to ferrylHb in atherosclerotic plaques exhibited a proinflammatory phenotype, as reflected by elevated levels of IL-1β and TNF-α. To find further signatures of ferrylHb in complicated lesions, we performed RNA-seq analysis on biopsies from patients who underwent endarterectomies. RNA-seq analysis demonstrated that human complicated lesions had a unique transcriptomic profile different from arteries and atheromatous plaques. Pathways affected in complicated lesions included gene changes associated with phosphoinositide 3-kinase (PI3K) signaling, lipid transport, tissue remodeling, and vascularization. Targeted analysis of gene expression associated with calcification, apoptosis, and hemolytic-specific clusters indicated an increase in the severity of complicated lesions compared with atheroma. A 39% overlap in the differential gene expression profiles of human macrophages exposed to ferrylHb and the complicated lesion profiles was uncovered. Among these 547 genes, we found inflammatory, angiogenesis, and iron metabolism gene clusters regulated in macrophages. Innovation and Conclusion: We conclude that oxidation of Hb to ferrylHb contributes to the progression of atherosclerosis via polarizing macrophages into a proatherogenic phenotype. Antioxid. Redox Signal. 35, 917-950.
Collapse
Affiliation(s)
- László Potor
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Zoltán Hendrik
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary.,Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andreas Patsalos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Medicine and Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA.,Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Éva Katona
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilárd Póliska
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gergő Kalló
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Komáromi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Combi
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Niké Posta
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Katalin Éva Sikura
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dávid Pethő
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Melinda Oros
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Tóth
- Division of Vascular Surgery, Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Gergely
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Medicine and Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - György Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
16
|
Kang H, Bienzle D, Lee GKC, Piché É, Viel L, Odemuyiwa SO, Beeler-Marfisi J. Flow cytometric analysis of equine bronchoalveolar lavage fluid cells in horses with and without severe equine asthma. Vet Pathol 2021; 59:91-99. [PMID: 34521286 PMCID: PMC8679174 DOI: 10.1177/03009858211042588] [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] [Indexed: 01/09/2023]
Abstract
Severe equine asthma (SEA) is a common, debilitating lower airway inflammatory
disorder of older horses. Alveolar macrophages (AMs) survey inhaled particulates
from barn sources causing them to switch from an anti-inflammatory to a
proinflammatory phenotype, resulting in neutrophil recruitment to the lung. This
proinflammatory switch may contribute to the development and prolongation of
SEA. Validated antibodies to identify the cells involved in the pathogenesis of
SEA are lacking. In this study, monoclonal antibodies against CD90, CD163, and
CD206 were tested for reactivity with equine leukocytes by immunocytochemistry
and flow cytometry. A multi-color flow cytometric assay was developed to
identify leukocytes in equine bronchoalveolar lavage fluid (BALF). Four control
and 4 SEA-susceptible horses had BALF collected before and after a 48-hour moldy
hay challenge. Antibodies against CD90 uniquely labeled equine neutrophils, and
antibodies against CD163 and CD206 identified equine macrophages. Postchallenge
AM surface expression of CD163 increased in both groups of horses, but the
increase was statistically significant in only the SEA-susceptible group
(P = .02). The surface expression of CD206 on AMs increased
significantly in the SEA-susceptible group (P = .03) but was
unchanged in the control group (P = .5). Increased expression
of CD163 and CD206 during exacerbation of SEA suggested an association between
AM phenotype and lung inflammation. However, functions of AMs in the
pathogenesis of SEA remain to be elucidated.
Collapse
Affiliation(s)
- Heng Kang
- University of Guelph, Guelph, Ontario, Canada
| | | | | | - Érica Piché
- University of Guelph, Guelph, Ontario, Canada
| | | | | | | |
Collapse
|
17
|
Milenova I, Lopez Gonzalez M, Quixabeira DCA, Santos JM, Cervera-Carrascon V, Dong W, Hemminki A, van Beusechem VW, van de Ven R, de Gruijl TD. Oncolytic Adenovirus ORCA-010 Activates Proinflammatory Myeloid Cells and Facilitates T Cell Recruitment and Activation by PD-1 Blockade in Melanoma. Hum Gene Ther 2021; 32:178-191. [PMID: 33470166 DOI: 10.1089/hum.2020.277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors have advanced the treatment of melanoma. Nevertheless, a majority of patients are resistant, or develop resistance, to immune checkpoint blockade, which may be related to prevailing immune suppression by myeloid regulatory cells in the tumor microenvironment (TME). ORCA-010 is a novel oncolytic adenovirus that selectively replicates in, and lyses, cancer cells. We previously showed that ORCA-010 can activate melanoma-exposed conventional dendritic cells (cDCs). To study the effect of ORCA-010 on melanoma-conditioned macrophage development, we used an in vitro co-culture model of human monocytes with melanoma cell lines. We observed a selective survival and polarization of monocytes into M2-like macrophages (CD14+CD80-CD163+) in co-cultures with cell lines that expressed macrophage colony-stimulating factor. Oncolysis of these melanoma cell lines, effected by ORCA-010, activated the resulting macrophages and converted them to a more proinflammatory state, evidenced by higher levels of PD-L1, CD80, and CD86 and an enhanced capacity to prime allogenic T cells and induce a type-1 T cell response. To assess the effect of ORCA-010 on myeloid subset distribution and activation in vivo, ORCA-010 was intratumorally injected and tested for T cell activation and recruitment in the human adenovirus nonpermissive B16-OVA mouse melanoma model. While systemic PD-1 blockade in this model in itself did not modulate myeloid or T cell subset distribution and activation, when it was preceded by i.t. injection of ORCA-010, this induced an increased rate and activation state of CD8α+ cDC1, both in the TME and in the spleen. Observed increased rates of activated CD8+ T cells, expressing CD69 and PD-1, were related to both increased CD8α+ cDC1 rates and M1/M2 shifts in tumor and spleen. In conclusion, the myeloid modulatory properties of ORCA-010 in melanoma, resulting in recruitment and activation of T cells, could enhance the antitumor efficacy of PD-1 blockade.
Collapse
Affiliation(s)
- Ioanna Milenova
- Departments of Medical Oncology and Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands.,ORCA Therapeutics BV, 's-Hertogenbosch, The Netherlands
| | - Marta Lopez Gonzalez
- Departments of Medical Oncology and Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Dafne C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Joao Manuel Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wenliang Dong
- ORCA Therapeutics BV, 's-Hertogenbosch, The Netherlands
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Victor W van Beusechem
- Departments of Medical Oncology and Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Departments of Medical Oncology and Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands.,Departments of Otolaryngology/Head-Neck Surgery, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Departments of Medical Oncology and Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Jiang W, Luo Z, Stephenson S, Li H, Di Germanio C, Norris PJ, Fuchs D, Zetterberg H, Gisslen M, Price RW. Cerebrospinal Fluid and Plasma Lipopolysaccharide Levels in Human Immunodeficiency Virus Type 1 Infection and Associations With Inflammation, Blood-Brain Barrier Permeability, and Neuronal Injury. J Infect Dis 2020; 223:1612-1620. [PMID: 33320240 DOI: 10.1093/infdis/jiaa765] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
Human immunodeficiency virus (HIV) infection is associated with increased systemic microbial translocation, neuroinflammation, and occasionally, neuronal injury. Whether systemic lipopolysaccharide (LPS) penetrates into the brain and contributes to neuroinflammation remain unknown in HIV. Here, we measured plasma and cerebrospinal fluid (CSF) LPS levels along with biomarkers of neuroinflammation (white blood cell counts and 40 soluble markers) and neurofilament light chain (NfL). Notably, CSF LPS was undetectable in all samples, including 3 HIV-infected individuals with dementia. Increased plasma LPS, neuroinflammation, and blood-brain barrier (BBB) dysfunction were found in untreated HIV-infected individuals, but not in healthy or treated HIV-infected individuals. Plasma LPS levels were directly correlated with various markers of inflammation in both plasma and CSF, as well as with degree of BBB permeability but not with CSF NfL in HIV-infected subjects. These results suggest that the magnitude of microbial translocation associates with neuroinflammation and BBB permeability in HIV without direct penetration into the central nervous system.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA.,Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sophie Stephenson
- Department of Neurology, University of California, San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Hong Li
- Public Health Sciences, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | - Dietmar Fuchs
- Institut für Biologische Chemie, Biozentrum, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London, United Kingdom.,United Kingdom Dementia Research Institute at University College London, London, United Kingdom
| | - Magnus Gisslen
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Diseases, Gothenburg, Sweden
| | - Richard W Price
- Department of Neurology, University of California, San Francisco, San Francisco General Hospital, San Francisco, California, USA
| |
Collapse
|
19
|
Fujiwara Y, Ohnishi K, Horlad H, Saito Y, Shiraishi D, Takeya H, Yoshii D, Kaieda S, Hoshino T, Komohara Y. CD163 deficiency facilitates lipopolysaccharide-induced inflammatory responses and endotoxin shock in mice. Clin Transl Immunology 2020; 9:e1162. [PMID: 33005412 PMCID: PMC7518957 DOI: 10.1002/cti2.1162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/12/2022] Open
Abstract
Objectives Septic (or endotoxin) shock is a severe systemic inflammatory disease caused by bacteraemia or endotoxaemia. Although it is known that increased serum levels of CD163 are observed in septic/endotoxin shock patients, the exact function and significance of CD163 in macrophage activation remain unclear. Therefore, in the current study, we tested whether CD163 contributes to the pathogenesis of endotoxin shock in mice. Methods and results In samples obtained from autopsy, the number of CD163‐positive macrophages was increased in the kidney, liver, heart, bone marrow and spleen of patients who had died from septic/endotoxin shock when compared to patients who had died from other causes. The animal study revealed a significantly lower survival rate in CD163‐deficient mice after lipopolysaccharide (LPS) injection. Several cytokines and oxidative stress‐related molecules were significantly elevated in the sera of LPS‐induced endotoxin shock mice models. Higher concentrations of IL‐6, TNF‐α, IL‐1β, nitrite (NO2‐) and nitrate (NO3‐) and a lower concentration of IL‐10 were observed in CD163‐deficient mice treated with LPS. Similar results were observed in CD163‐deficient LPS‐stimulated macrophages. Furthermore, in an antitype II collagen antibody‐induced arthritis (CAIA), rheumatoid arthritis model, inflammation and bone erosion scores as well as the expression of IL‐6 and IL‐1β were significantly increased in CD163‐deficient mice. Conclusions CD163 was suggested to be involved in the regulation of inflammatory cytokine expression in septic/endotoxin shock and CAIA.
Collapse
Affiliation(s)
- Yukio Fujiwara
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Koji Ohnishi
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Hasita Horlad
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Yoichi Saito
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan.,Laboratory of Biomaterials Institute for Frontier Life and Medical Sciences Kyoto University Kyoto Japan.,Research Fellow of Japan Society for the Promotion of Science Tokyo Japan
| | - Daisuke Shiraishi
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Hiroto Takeya
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Daiki Yoshii
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Shinjiro Kaieda
- Division of Respirology, Neurology, and Rheumatology Department of Medicine Kurume University School of Medicine Kurume Japan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology Department of Medicine Kurume University School of Medicine Kurume Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan.,Center for Metabolic Regulation of Healthy Aging Kumamoto University Kumamoto Japan
| |
Collapse
|
20
|
Kraaijvanger R, Janssen Bonás M, Vorselaars ADM, Veltkamp M. Biomarkers in the Diagnosis and Prognosis of Sarcoidosis: Current Use and Future Prospects. Front Immunol 2020; 11:1443. [PMID: 32760396 PMCID: PMC7372102 DOI: 10.3389/fimmu.2020.01443] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Sarcoidosis is a heterogeneous disease in terms of presentation, duration, and severity. Due to this heterogeneity, it is difficult to align treatment decisions. Biomarkers have proved to be useful for the diagnosis and prognosis of many diseases, and over the years, many biomarkers have been proposed to facilitate diagnosis, prognosis, and treatment decisions. Unfortunately, the ideal biomarker for sarcoidosis has not yet been discovered. The most commonly used biomarkers are serum and bronchoalveolar lavage biomarkers, but these lack the necessary specificity and sensitivity. In sarcoidosis, therefore, a combination of these biomarkers is often used to establish a proper diagnosis or detect possible progression. Other potential biomarkers include imaging tools and cell signaling pathways. Fluor-18-deoxyglucose positron emission tomography and high-resolution computed tomography have been proven to be more sensitive for the diagnosis and prognosis of both pulmonary and cardiac sarcoidosis than the serum biomarkers ACE and sIL-2R. There is an upcoming role for exploration of signaling pathways in sarcoidosis pathogenesis. The JAK/STAT and mTOR pathways in particular have been investigated because of their role in granuloma formation. The activation of these signaling pathways also proved to be a specific biomarker for the prognosis of sarcoidosis. Furthermore, both imaging and cell signaling biomarkers also enable patients who might benefit from a particular type of treatment to be distinguished from those who will not. In conclusion, the diagnostic and prognostic path of sarcoidosis involves many different types of existing and new biomarker. Research addressing biomarkers and disease pathology is ongoing in order to find the ideal sensitive and specific biomarker for this disease.
Collapse
Affiliation(s)
- Raisa Kraaijvanger
- Department of Pulmonology, ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Montse Janssen Bonás
- Department of Pulmonology, ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Adriane D. M. Vorselaars
- Department of Pulmonology, ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Marcel Veltkamp
- Department of Pulmonology, ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, Netherlands
- Department of Pulmonology, University Medical Center, Utrecht, Netherlands
| |
Collapse
|
21
|
Vakili S, Ahooyi TM, Yarandi SS, Donadoni M, Rappaport J, Sariyer IK. Molecular and Cellular Impact of Inflammatory Extracellular Vesicles (EVs) Derived from M1 and M2 Macrophages on Neural Action Potentials. Brain Sci 2020; 10:brainsci10070424. [PMID: 32635207 PMCID: PMC7408497 DOI: 10.3390/brainsci10070424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022] Open
Abstract
Several factors can contribute to neuroinflammatory disorders, such as cytokine and chemokines that are produced and released from peripherally derived immune cells or from locally activated cells such as microglia and perivascular macrophages in the brain. The primary function of these cells is to clear inflammation; however, following inflammation, circulating monocytes are recruited to the central nervous system (CNS). Monocyte-derived macrophages in the CNS play pivotal roles in mediating neuroinflammatory responses. Macrophages are heterogeneous both in normal and in pathological conditions due to their plasticity, and they are classified in two main subsets, classically activated (M1) or alternatively activated (M2). There is accumulating evidence suggesting that extracellular vesicles (EVs) released from activated immune cells may play crucial roles in mediating inflammation. However, a possible role of EVs released from immune cells such as M1 and M2 macrophages on neuronal functions in the brain is not known. In order to investigate the molecular and cellular impacts of macrophages and EVs released from macrophage subtypes on neuronal functions, we used a recently established in vitro M1 and M2 macrophage culture model and isolated and characterized EVs from these macrophage subtypes, treated primary neurons with M1 or M2 EVs, and analyzed the extracellular action potentials of neurons with microelectrode array studies (MEA). Our results introduce evidence on the interfering role of inflammatory EVs released from macrophages in interneuronal signal transmission processes, with implications in the pathogenesis of neuroinflammatory diseases induced by a variety of inflammatory insults.
Collapse
Affiliation(s)
- Sarah Vakili
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (S.V.); (T.M.A.); (S.S.Y.); (M.D.)
| | - Taha Mohseni Ahooyi
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (S.V.); (T.M.A.); (S.S.Y.); (M.D.)
| | - Shadan S. Yarandi
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (S.V.); (T.M.A.); (S.S.Y.); (M.D.)
| | - Martina Donadoni
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (S.V.); (T.M.A.); (S.S.Y.); (M.D.)
| | - Jay Rappaport
- Tulane National Primate Research Center, New Orleans, Covington, LA 70433, USA;
| | - Ilker K. Sariyer
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (S.V.); (T.M.A.); (S.S.Y.); (M.D.)
- Correspondence:
| |
Collapse
|
22
|
Vakili S, Fischer T, Rappaport J. M2 differentiation of MonoMac‐1 cell line induced by M‐CSF and glucocorticoid pathways. J Cell Physiol 2020; 235:7383-7391. [DOI: 10.1002/jcp.29638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/15/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Sarah Vakili
- Center for Neurovirology, Department of NeuroscienceTemple University School of MedicinePhiladelphia Pennsylvania
| | - Tracy Fischer
- Center for Neurovirology, Department of NeuroscienceTemple University School of MedicinePhiladelphia Pennsylvania
| | - Jay Rappaport
- Center for Neurovirology, Department of NeuroscienceTemple University School of MedicinePhiladelphia Pennsylvania
| |
Collapse
|
23
|
Underwood ML, Nguyen T, Uebelhoer LS, Kunkel LE, Korthuis PT, Lancioni CL. Altered monocyte phenotype and dysregulated innate cytokine responses among people living with HIV and opioid-use disorder. AIDS 2020; 34:177-188. [PMID: 31687981 PMCID: PMC6948804 DOI: 10.1097/qad.0000000000002416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Supplemental Digital Content is available in the text Background: Opioid-use disorders (OUD) and hepatitis C or B co-infection (HEP) are common among people living with HIV (PLHIV). The impact of OUD on innate and adaptive immunity among PLHIV with and without HEP is unknown. Objectives: To investigate the impact of OUD on monocyte and T-cell phenotypes, cytokine responses to lipopolysaccharide (LPS) and phytohemagglutinin (PHA), and plasma inflammatory markers, among PLHIV with and without HEP. Methods: Cross-sectional study enrolling PLHIV receiving ART, with and without OUD. Flow cytometry determined monocyte and T-cell phenotypes; LPS and PHA-induced cytokine production was assessed following LPS and PHA stimulation by multiplex cytokine array; plasma IL-6, soluble CD163, and soluble CD14 were measured by ELISA. Results: Twenty-two PLHIV with OUD and 37 PLHIV without OUD were included. PLHIV with OUD exhibited higher frequencies of intermediate (CD14++CD16+) and nonclassical (CD14dimCD16+) monocytes when compared with PLHIV without OUD (P = 0.0025; P = 0.0001, respectively), regardless of HEP co-infection. Soluble CD163 and monocyte cell surface CD163 expression was increased among PLHIV with OUD and HEP, specifically. Regardless of HEP co-infection, PLHIV with OUD exhibited reduced production of IL-10, IL-8, IL-6, IL-1alpha, and TNF-alpha in response to LPS when compared with PLHIV without OUD; PHA-induced production of IL-10, IL-1alpha, IL-1beta, IL-6, and TNF-alpha were also reduced among individuals with OUD. Conclusion: OUD among PLHIV are associated with altered monocyte phenotypes and a dysregulated innate cytokine response. Defining underlying mechanisms of opioid-associated innate immune dysregulation among PLHIV should be prioritized to identify optimal OUD treatment strategies.
Collapse
|
24
|
High Plasma Soluble CD163 During Infancy Is a Marker for Neurocognitive Outcomes in Early-Treated HIV-Infected Children. J Acquir Immune Defic Syndr 2019; 81:102-109. [PMID: 30768490 DOI: 10.1097/qai.0000000000001979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Monocyte activation may contribute to neuronal injury in aviremic HIV-infected adults; data are lacking in children. We examined the relation between monocyte activation markers and early and long-term neurodevelopmental outcomes in early-treated HIV-infected children. SETTING Prospective study of infant and child neurodevelopmental outcomes nested within a randomized clinical trial (NCT00428116) and extended cohort study in Kenya. METHODS HIV-infected infants (N = 67) initiated antiretroviral therapy (ART) at age <5 months. Plasma soluble (s) CD163 (sCD163), sCD14, and neopterin were measured before ART (entry) and 6 months later. Milestone attainment was ascertained monthly during 24 months, and neuropsychological tests were performed at 5.8-8.2 years after initiation of ART (N = 27). The relationship between neurodevelopment and sCD163, sCD14, and neopterin at entry and 6 months after ART was assessed using Cox proportional hazards models and linear regression. RESULTS Infants with high entry sCD163 had unexpected earlier attainment of supported sitting (5 vs 6 months; P = 0.006) and supported walking (10 vs 12 months; P = 0.02) with trends in adjusted analysis. Infants with high 6-month post-ART sCD163 attained speech later (17 vs 15 months; P = 0.006; adjusted hazard ratio, 0.47; P = 0.02), threw toys later (18 vs 17 months; P = 0.01; adjusted hazard ratio, 0.53; P = 0.04), and at median 6.8 years after ART, had worse neuropsychological test scores (adj. mean Z-score differences, cognition, -0.42; P = 0.07; short-term memory, -0.52; P = 0.08; nonverbal test performance, -0.39, P = 0.05). CONCLUSIONS Before ART, monocyte activation may reflect transient neuroprotective mechanisms in infants. After ART and viral suppression, monocyte activation may predict worse short- and long-term neurodevelopment outcomes.
Collapse
|
25
|
Huang X, Li Y, Guo X, Zhu Z, Kong X, Yu F, Wang Q. Identification of differentially expressed genes and signaling pathways in chronic obstructive pulmonary disease via bioinformatic analysis. FEBS Open Bio 2019; 9:1880-1899. [PMID: 31419078 PMCID: PMC6823288 DOI: 10.1002/2211-5463.12719] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/07/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a multifactorial and heterogeneous disease that creates public health challenges worldwide. The underlying molecular mechanisms of COPD are not entirely clear. In this study, we aimed to identify the critical genes and potential molecular mechanisms of COPD by bioinformatic analysis. The gene expression profiles of lung tissues of COPD cases and healthy control subjects were obtained from the Gene Expression Omnibus. Differentially expressed genes were analyzed by integration with annotations from Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, followed by construction of a protein‐protein interaction network and weighted gene coexpression analysis. We identified 139 differentially expressed genes associated with the progression of COPD, among which 14 Hub genes were identified and found to be enriched in certain categories, including immune and inflammatory response, response to lipopolysaccharide and receptor for advanced glycation end products binding; in addition, these Hub genes are involved in multiple signaling pathways, particularly hematopoietic cell lineage and cytokine‐cytokine receptor interaction. The 14 Hub genes were positively or negatively associated with COPD by wgcna analysis. The genes CX3CR1,PTGS2,FPR1,FPR2, S100A12,EGR1,CD163, S100A8 and S100A9 were identified to mediate inflammation and injury of the lung, and play critical roles in the pathogenesis of COPD. These findings improve our understanding of the underlying molecular mechanisms of COPD.
Collapse
Affiliation(s)
- Xinwei Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, China.,Medical School, Kunming University of Science and Technology, China
| | - Yunwei Li
- Medical School, Kunming University of Science and Technology, China.,Department of Pharmacy, Kunming Children's Hospital, China
| | - Xiaoran Guo
- Medical School, Kunming University of Science and Technology, China
| | - Zongxin Zhu
- Medical School, Kunming University of Science and Technology, China
| | - Xiangyang Kong
- Medical School, Kunming University of Science and Technology, China
| | - Fubing Yu
- Department of Gastroenterology, Fourth Affiliated Hospital of Kunming Medical University, China
| | - Qiang Wang
- Physical Examination Center, Second People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
26
|
Nouno T, Okamoto M, Ohnishi K, Kaieda S, Tominaga M, Zaizen Y, Ichiki M, Momosaki S, Nakamura M, Fujimoto K, Fukuoka J, Shimizu S, Komohara Y, Hoshino T. Elevation of pulmonary CD163 + and CD204 + macrophages is associated with the clinical course of idiopathic pulmonary fibrosis patients. J Thorac Dis 2019; 11:4005-4017. [PMID: 31656675 DOI: 10.21037/jtd.2019.09.03] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background M2-like/repair macrophages are thought to contribute to fibrotic process of idiopathic pulmonary fibrosis (IPF). We analyzed the association between pulmonary accumulation of M2-like macrophages and survival in IPF patients. Methods Lung tissues were obtained by surgical lung biopsy from patients with IPF (n=16), nonspecific interstitial pneumonia (NSIP, n=8) and control subjects (n=14). Samples were also obtained at autopsy from 9 patients who died of acute exacerbation (AE) of IPF. Lung specimens and/or human peripheral blood mononuclear cells-derived macrophages were evaluated by immunohistochemistry for expression of CD68 (pan-macrophage marker), CD163, and CD204 (M2-like macrophage markers), and by in situ mRNA hybridization and ELISA for production of transforming growth factor-β1 (TGF-β1). Results CD68+, CD163+, and CD204+ cell counts and CD163+/CD68+ and CD204+/CD68+ cell ratios were comparable in IPF and NSIP lung tissues and significantly higher than in control tissues. IPF-AE lung samples contained significantly elevated CD68+ and CD163+ cell counts and CD163+/CD68+ cell ratio compared with IPF samples, whereas CD204+ cell counts and CD204+/CD68+ cells ratio did not differ. High CD163+/CD68+ and CD204+/CD68+ cell ratios were significantly associated with shorter overall survival and time-to-AE in IPF patients. In vitro-differentiated human CD163+ and CD204+ macrophages both secreted TGF-β1; however, the novel IPF drug pentraxin 2/serum amyloid protein could suppress secretion only by CD204+ macrophages. Conclusions Pulmonary accumulation of CD163+ and CD204+ macrophages is associated with worse clinical course in IPF patients. Suppression of macrophage activation and TGF-β1 secretion may be a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Takashi Nouno
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| | - Masaki Okamoto
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuou-ku, Kumamoto, Japan
| | - Shinjiro Kaieda
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| | - Masaki Tominaga
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| | - Yoshiaki Zaizen
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| | - Masao Ichiki
- Department of Respirology, National Hospital Organization Kyushu Medical Center, Jigyohama, Chuou-ku, Fukuoka, Japan
| | - Seiya Momosaki
- Department of Pathology, National Hospital Organization Kyushu Medical Center, Jigyohama, Chuou-ku, Fukuoka, Japan
| | - Masayuki Nakamura
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| | - Kiminori Fujimoto
- Department of Radiology and Center for Diagnostic Imaging, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| | - Junya Fukuoka
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto, Nagasaki, Japan
| | - Shigeki Shimizu
- Department of Pathology, Kindai University Faculty of Medicine, Ohnohigashi, Osakasayama, Osaka, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuou-ku, Kumamoto, Japan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Asahi-machi, Kurume, Japan
| |
Collapse
|
27
|
Montoya D, Mehta M, Ferguson BG, Teles RMB, Krutzik SR, Cruz D, Pellegrini M, Modlin RL. Plasticity of antimicrobial and phagocytic programs in human macrophages. Immunology 2019; 156:164-173. [PMID: 30357820 PMCID: PMC6328994 DOI: 10.1111/imm.13013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/01/2018] [Indexed: 01/03/2023] Open
Abstract
Macrophage (MΦ) polarization is triggered during the innate immune response to defend against microbial pathogens, but can also contribute to disease pathogenesis. In a previous study, we found that interleukin-15 (IL-15) -derived classically activated macrophages (M1 MΦ) have enhanced antimicrobial activity, whereas IL-10-derived alternatively activated macrophages (M2 MΦ) were highly phagocytic but lacked antimicrobial activity. Given that the ability to modulate MΦ polarization from M2 MΦ to M1 MΦ may promote a more effective immune response to infection, we investigated the plasticity of these MΦ programs. Addition of IL-10 to M1 MΦ induced M2-like MΦ, but IL-15 had little effect on M2 MΦ. We determined the set of immune receptors that are present on M2 MΦ, elucidating two candidates for inducing plasticity of M2 MΦ, Toll-like receptor 1 (TLR1) and interferonγ (IFN-γ) receptor 1. Stimulation of M2 MΦ with TLR2/1 ligand (TLR2/1L) or IFN-γ alone was not sufficient to alter M2 MΦ phenotype or function. However, co-addition of TLR2/1L and IFN-γ re-educated M2 MΦ towards the M1 MΦ phenotype, with a decrease in the phagocytosis of lipids and mycobacteria, as well as recovery of the vitamin-D-dependent antimicrobial pathway compared with M2 MΦ maintained in polarizing conditions. Similarly, treatment of M2 MΦ with both TLR2/1L and anti-IL-10 neutralizing antibodies led to polarization to the M1-like MΦ phenotype and function. Together, our data demonstrate an approach to induce MΦ plasticity that provides the potential for re-educating MΦ function in human mycobacterial disease to promote host defense and limit pathogenesis.
Collapse
Affiliation(s)
- Dennis Montoya
- Department of Molecular, Cell, and Developmental BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Manali Mehta
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | | | - Rosane M. B. Teles
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Stephan R. Krutzik
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Daniel Cruz
- Division of CardiologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Robert L. Modlin
- Division of DermatologyDepartment of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
28
|
Abstract
Viral infection in the brain can be acute or chronic, with the responses often producing foci of increasingly cytotoxic inflammation. This can lead to effects beyond the central nervous system (CNS). To stimulate discussion, this commentary addresses four questions: What drives the development of human immunodeficiency virus (HIV)-associated neurocognitive disorders, does the phenotype of macrophages in the CNS spur development of HIV encephalitis (HIVE), does continual activation of astrocytes drive the development of HIV-associated neurocognitive disorders/subclinical disease, and neuroinflammation: friend or foe? A unifying theory that connects each question is the issue of continued activation of glial cells, even in the apparent absence of simian immunodeficiency virus/HIV in the CNS. As the CNS innate immune system is distinct from the rest of the body, it is likely there could be a number of activation profiles not observed elsewhere.
Collapse
Affiliation(s)
- Elizabeth C. Delery
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane Program in Biomedical Sciences, Tulane Medical School, New Orleans, Louisiana
- Department of Microbiology and Immunology, Tulane Medical School, New Orleans, Louisiana
| | - Andrew G. MacLean
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane Program in Biomedical Sciences, Tulane Medical School, New Orleans, Louisiana
- Department of Microbiology and Immunology, Tulane Medical School, New Orleans, Louisiana
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana
- Center for Aging, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
29
|
Ding D, Yao Y, Yang C, Zhang S. Identification of mannose receptor and CD163 as novel biomarkers for colorectal cancer. Cancer Biomark 2018; 21:689-700. [PMID: 29226859 DOI: 10.3233/cbm-170796] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Dongbing Ding
- Department of Gastrointestinal Surgery, Jingmen First People’s Hospital, Jingmen 448000, Hubei, China
- Department of Gastrointestinal Surgery, Jingmen First People’s Hospital, Jingmen 448000, Hubei, China
| | - Yao Yao
- Department of Ophthalmology, Jingmen First People’s Hospital, Jingmen 448000, Hubei, China
- Department of Gastrointestinal Surgery, Jingmen First People’s Hospital, Jingmen 448000, Hubei, China
| | - Changming Yang
- Department of Anesthesiology, Jingmen First People’s Hospital, Jingmen 448000, Hubei, China
| | - Songbai Zhang
- Department of Gastrointestinal Surgery, Jingmen First People’s Hospital, Jingmen 448000, Hubei, China
| |
Collapse
|
30
|
Impaired Inflammatory Response to LPS in Type 2 Diabetes Mellitus. Int J Inflam 2018; 2018:2157434. [PMID: 29568481 PMCID: PMC5820544 DOI: 10.1155/2018/2157434] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a severe health problem worldwide, reaching epidemic levels. High susceptibility to infections of T2DM patients indicates dysregulated immune responses to pathogens. However, innate immune responses, including monocyte functions, in T2DM are poorly investigated. Therefore, in this study we aimed to assess lipopolysaccharide- (LPS-) induced immune responses of circulating monocytes from T2DM patients. The results showed that monocytes from T2DM were hyporesponsive to LPS challenge as reflected by significantly suppressed secretion of TNFα (p < 0.01) and expression of CD11b (p < 0.001) and TLR4 (p < 0.001) compared to those in monocytes from healthy subjects. Furthermore, LPS-induced IL-10 levels were similar in diabetic and healthy supernatants, while expression levels of CD163 were found to be downregulated on monocytes from T2DM (p < 0.001) suggesting impaired ability of monocytes to switch their phenotype to anti-inflammatory. Taken together, our results suggest compromised function of monocytes in T2DM, which may explain, at least partly, high incidence of infection in these patients.
Collapse
|
31
|
Shamaei M, Mortaz E, Pourabdollah M, Garssen J, Tabarsi P, Velayati A, Adcock IM. Evidence for M2 macrophages in granulomas from pulmonary sarcoidosis: A new aspect of macrophage heterogeneity. Hum Immunol 2017; 79:63-69. [PMID: 29107084 DOI: 10.1016/j.humimm.2017.10.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/11/2017] [Accepted: 10/24/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Sarcoidosis is a granulomatous disease of unknown etiology. Macrophages play a key role in granuloma formation with the T cells, having a significant impact on macrophage polarization (M1 and M2) and the cellular composition of the granuloma. This study evaluates macrophage polarization in granulomas in pulmonary sarcoidosis. MATERIALS AND METHODS Tissue specimens from the Department of Pathology biobank at the Masih Daneshvari Hospital were obtained. Paraffin sections from 10 sarcoidosis patients were compared with those from 12 cases of tuberculosis using immunohistochemical staining. These sections consisted of mediastinal lymph nodes and transbronchial lung biopsy (TBLB) for sarcoidosis patients versus pleural tissue, neck, axillary lymph nodes and TBLB for tuberculosis patients. The sections were stained for T-cells (CD4+, CD8+) and mature B lymphocytes (CD22+). CD14+ and CD68+ staining was used as a marker of M1 macrophages and CD163+ as a marker for M2 macrophages. RESULTS Immunohistochemical staining revealed a 4/1 ratio of CD4+/CD8+ T-cells in sarcoidosis granuloma sections and a 3/1 ratio in tuberculosis sections. There was no significance difference in single CD4+, CD8+, CD22+, CD14+ and CD68+ staining between sarcoidosis and tuberculosis sections. CD163 expression was significantly increased in sarcoidosis sections compared with those from tuberculosis subjects. CONCLUSION Enhanced CD163+ staining indicates a shift towards M2 macrophage subsets in granulomas from sarcoidosis patients. Further research is required to determine the functional role of M2 macrophages in the immunopathogenesis of sarcoidosis.
Collapse
Affiliation(s)
- Masoud Shamaei
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Mihan Pourabdollah
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Nutricia Research Centre for Specialized Nutrition, Utrecht, Netherlands
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aliakbar Velayati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M Adcock
- Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, UK; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
32
|
Ding D, Song Y, Yao Y, Zhang S. Preoperative serum macrophage activated biomarkers soluble mannose receptor (sMR) and soluble haemoglobin scavenger receptor (sCD163), as novel markers for the diagnosis and prognosis of gastric cancer. Oncol Lett 2017; 14:2982-2990. [PMID: 28928836 PMCID: PMC5588128 DOI: 10.3892/ol.2017.6547] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/06/2017] [Indexed: 12/15/2022] Open
Abstract
Soluble mannose receptor (sMR) and soluble haemoglobin scavenger receptor (sCD163) are macrophage activation markers which have previously been demonstrated to be increased in patients with inflammation, auto-immunity and malignancies. To investigate the clinical diagnostic and prognostic significance of preoperative serum sMR and sCD163, the present study investigated 143 gastric cancer (GC) patients, 66 subjects with benign gastric disease and 59 healthy controls, using an ELISA assay. Preoperative serum levels of sMR and sCD163 ranged from 0.165 to 0.885 µg/ml (median=0.374 µg/ml) and from 0.291 to 1.760 µg/ml (median=0.628 µg/ml) in GC patients, respectively. The expression levels of sMR and sCD163 were elevated compared with all controls (P<0.0001). Receiver operating characteristic analyses suggested that the optimum diagnostic cut-offs for sMR and sCD163 were 0.3405 µg/ml [area under curve (AUC) 0.7284, sensitivity 61.54%, and specificity 73.60%] and 0.6645 µg/ml (AUC 0.7766, sensitivity 53.85%, and specificity 86.40%), respectively. Notably, the measurement of serum sMR and sCD163 levels in conjugation, markedly enhanced the diagnostic accuracy (AUC 0.8490, sensitivity 70.63% and specificity 84.00%). Preoperative serum sMR and sCD163 levels correlated significantly with serum carcinoembryonic antigen, CA199, CA724 and CA125 concentrations in GC patients (P<0.05), however this association was not observed with sMR and CA724. High preoperative serum sMR and sCD163 levels correlated significantly with shorter overall survival (P=0.0041; P<0.0001, respectively) and were demonstrated to act as adverse prognostic factors (P=0.006; P<0.001, respectively). Furthermore, preoperative serum sMR and sCD163 levels correlated positively with the degree of lymphatic and distant metastasis of GC. In conclusion, preoperative serum sMR and sCD163 may be novel diagnostic and prognostic markers for GC and further studies are required in order to elucidate the underlying molecular mechanisms of sMR and CD163 in the development and progression of GC.
Collapse
Affiliation(s)
- Dongbing Ding
- Department of Gastrointestinal Surgery, Jingmen First People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Yang Song
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, P.R. China
| | - Yao Yao
- Department of Ophthalmology, Jingmen First People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Songbai Zhang
- Department of Gastrointestinal Surgery, Jingmen First People's Hospital, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
33
|
Jain S, Bose A, Bastia B, Sharma H, Sachdeva R, Jain AK, Pal R. Oxidized Hemoglobin Is Antigenic and Immunogenic in Lupus. Front Immunol 2017; 8:732. [PMID: 28694810 PMCID: PMC5483465 DOI: 10.3389/fimmu.2017.00732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022] Open
Abstract
Hemolysis-associated anemia is characteristic of diseases such as atherosclerosis, lupus, malaria, and leishmaniasis; the toxic effects of free hemoglobin (Hb) have been extensively described. This study was based on the premise that release of this sequestered, inflammatory molecule can result in deleterious immunological consequences, particularly in the context of pre-existing lupus. IgG anti-Hb responses were detected in the sera of lupus patients. Lupus-prone mice exhibited heightened plasma Hb levels, and ferric (Fe3+) Hb triggered preferential release of lupus-associated cytokines from splenocytes derived from aging lupus-prone mice. Anti-Hb B cell precursor frequencies were heightened in such mice, which also expressed increased titers of anti-Hb antibodies in serum and in kidney eluates. Fe3+ Hb preferentially increased the functional maturation of bone marrow-derived dendritic cells (BMDCs) from lupus-prone mice, effects abrogated upon the inhibition of Stat3. Hb interacted with lupus-associated autoantigens extruded during apoptosis and coincubation of Hb and apoptotic blebs had additional maturation-inducing effects on lupus BMDCs. Immunization with Hb in lupus-prone mice induced antigen spreading to lupus-associated moieties; Hb-interacting autoantigens were preferentially targeted and increased complement deposition and glomerulosclerosis were observed. Hb therefore demonstrates both antigenicity and immunogenicity and triggers specific immuno-pathological effects in a lupus milieu.
Collapse
Affiliation(s)
- Sonia Jain
- Immunoendocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Anjali Bose
- Immunoendocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Banajit Bastia
- Division of Electron Microscopy, National Institute of Pathology-ICMR, New Delhi, India
| | - Hritika Sharma
- Immunoendocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Ruchi Sachdeva
- Immunoendocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Arun K Jain
- Division of Electron Microscopy, National Institute of Pathology-ICMR, New Delhi, India
| | - Rahul Pal
- Immunoendocrinology Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
34
|
Liu H, Lin D, Xiang H, Chen W, Zhao S, Peng H, Yang J, Chen P, Chen S, Lu H. The role of tumor necrosis factor-like weak inducer of apoptosis in atherosclerosis via its two different receptors. Exp Ther Med 2017; 14:891-897. [PMID: 28781615 DOI: 10.3892/etm.2017.4600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
At present, it is commonly accepted that atherosclerosis is a chronic inflammatory disease characterized by disorder of the arterial wall. As one of the inflammatory cytokines of the tumor necrosis factor superfamily, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) participates in the formation and progression of atherosclerosis. TWEAK, when binding to its initial receptor, fibroblast growth factor inducible molecule 14 (Fn14), exerts adverse biological functions in atherosclerosis, including dysfunction of endothelial cells, phenotypic change of smooth muscle cells and inflammatory responses of monocytes/macrophages. However, accumulating data supports that, besides Fn14, TWEAK also binds to cluster of differentiation (CD)163, an anti-inflammatory cytokine and a scavenger receptor exclusively expressed by monocytes and macrophages. Furthermore, it has been demonstrated that CD163 is able to internalize TWEAK and likely elicits protective effects in atherosclerosis by terminating inflammation induced by TWEAK. In the present study, the role of TWEAK in atherosclerosis was reviewed, with a predominant focus on CD163 and Fn14 receptors.
Collapse
Affiliation(s)
- Hengdao Liu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Dan Lin
- Qingdao Center for Disease Control and Prevention, Qingdao, Shandong 266033, P.R. China
| | - Hong Xiang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Wei Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shaoli Zhao
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hui Peng
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jie Yang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Pan Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shuhua Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hongwei Lu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
35
|
Detzen L, Chen SCY, Cheng B, Papapanou PN, Lalla E. Increased levels of soluble CD163 in periodontitis patients. J Clin Periodontol 2017; 44:585-590. [DOI: 10.1111/jcpe.12731] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2017] [Indexed: 11/28/2022]
Affiliation(s)
- Laurent Detzen
- Division of Periodontics; College of Dental Medicine; Columbia University; New York NY USA
- Orofacial Pathologies; Imaging and Biotherapies Laboratory; Faculty of Odontology; Paris Descartes University; Montrouge France
| | - Steven C. Y. Chen
- Division of Periodontics; College of Dental Medicine; Columbia University; New York NY USA
| | - Bin Cheng
- Department of Biostatistics; Mailman School of Public Health; Columbia University; New York NY USA
| | - Panos N. Papapanou
- Division of Periodontics; College of Dental Medicine; Columbia University; New York NY USA
| | - Evanthia Lalla
- Division of Periodontics; College of Dental Medicine; Columbia University; New York NY USA
| |
Collapse
|
36
|
Park M, Kim S, Adelman JS, Leon AE, Hawley DM, Dalloul RA. Identification and functional characterization of the house finch interleukin-1β. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 69:41-50. [PMID: 27998740 DOI: 10.1016/j.dci.2016.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 06/06/2023]
Abstract
Interleukin-1β (IL-1β), an inflammatory cytokine of the IL-1 family, is primarily produced as a precursor protein by monocytes and macrophages, then matures and becomes activated through proteolytic catalysis. Although the biological characteristics of avian IL-1β are well known, little information is available about its biological role in songbird species such as house finches that are vulnerable to naturally-occurring inflammatory diseases. In this study, house finch IL-1β (HfIL-1β) was cloned, expressed, and its biological function examined. Both precursor and mature forms of HfIL-1β consisting of 269 and 162 amino acids, respectively, were amplified from total RNA of spleen and cloned into expression vectors. HfIL-1β showed high sequential and tertiary structural similarity to chicken homologue that allowed detection of the expressed mature recombinant HfIL-1β (rHfIL-1β) with anti-ChIL-1β antibody by immunoblot analysis. For further characterization, we used primary splenocytes and hepatocytes that are predominant sources of IL-1β upon stimulation, as well as suitable targets to stimulation by IL-1β. Isolated house finch splenocytes were stimulated with rHfIL-1β in the presence and absence of concanavalin A (Con A), RNA was extracted and transcript levels of Th1/Th2 cytokines and a chemokine were measured by qRT-PCR. The addition of rHfIL-1β induced significant enhancement of IL-2 transcript, a Th1 cytokine, while transcription of IL-1β and the Th2 cytokine IL-10 was slightly enhanced by rHfIL-1β treatment. rHfIL-1β also led to elevated levels of the chemokine CXCL1 and nitric oxide production regardless of co-stimulation with Con A. In addition, the production of the acute phase protein serum amyloid A and the antimicrobial peptide LEAP2 was observed in HfIL-1β-stimulated hepatocytes. Taken together, these observations revealed the basic functions of HfIL-1β including the stimulatory effect on cell proliferation, production of Th1/Th2 cytokines and acute phase proteins by immune cells, thus providing valuable insight into how HfIL-1β is involved in regulating inflammatory response.
Collapse
Affiliation(s)
- Myeongseon Park
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Sungwon Kim
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA; The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - James S Adelman
- Department of Natural Resource Ecology and Management, Iowa State University, Ames, IA 50011, USA
| | - Ariel E Leon
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Dana M Hawley
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Rami A Dalloul
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
37
|
Diseases Primarily Affecting the Reproductive System. Vet Med (Auckl) 2017. [PMCID: PMC7150237 DOI: 10.1016/b978-0-7020-5246-0.00018-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Genetic resistance - an alternative for controlling PRRS? Porcine Health Manag 2016; 2:27. [PMID: 28405453 PMCID: PMC5382513 DOI: 10.1186/s40813-016-0045-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/19/2016] [Indexed: 12/22/2022] Open
Abstract
PRRS is one of the most challenging diseases for world-wide pig production. Attempts for a sustainable control of this scourge by vaccination have not yet fully satisfied. With an increasing knowledge and methodology in disease resistance, a new world-wide endeavour has been started to support the combat of animal diseases, based on the existence of valuable gene variants with regard to any host-pathogen interaction. Several groups have produced a wealth of evidence for natural variability in resistance/susceptibility to PRRS in our commercial breeding lines. However, up to now, exploiting existing variation has failed because of the difficulty to detect the carriers of favourable and unfavourable alleles, especially with regard to such complex polygenic traits like resistance to PRRS. New hope comes from new genomic tools like next generation sequencing which have become extremely fast and low priced. Thus, research is booming world-wide and the jigsaw puzzle is filling up – slowly but steadily. On the other hand, knowledge from virological and biomedical basic research has opened the way for an “intervening way”, i.e. the modification of identified key genes that occupy key positions in PRRS pathogenesis, like CD163. CD163 was identified as the striking receptor in PRRSV entry and its knockout from the genome by gene editing has led to the production of pigs that were completely resistant to PRRSV – a milestone in modern pig breeding. However, at this early step, concerns remain about the acceptance of societies for gene edited products and regulation still awaits upgrading to the new technology. Further questions arise with regard to upcoming patents from an ethical and legal point of view. Eventually, the importance of CD163 for homeostasis, defence and immunity demands for more insight before its complete or partial silencing can be answered. Whatever path will be followed, even a partial abolishment of PRRSV replication will lead to a significant improvement of the disastrous herd situation, with a significant impact on welfare, performance, antimicrobial consumption and consumer protection. Genetics will be part of a future solution.
Collapse
|
39
|
Onofre G, Koláčková M, Jankovičová K, Krejsek J. Scavenger Receptor CD163 and Its Biological Functions. ACTA MEDICA (HRADEC KRÁLOVÉ) 2016. [DOI: 10.14712/18059694.2016.105] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
CD163 is a member of scavenger receptor super family class B of the first subgroup. It is mapped to the region p13 on chromosome 12. Five different isoforms of CD163 have been described, which differ in the structure of their cytoplasmic domains and putative phosporylation sites. This scavenger receptor is selectively expressed on cells of monocytes and macrophages lineage exclusively. CD163 immunological function is essentially homeostatic. It also has other functions because participates in adhesion to endothelial cells, in tolerance induction and tissues regeneration. Other very important function of CD163 is the clearance of hemoglobin in its cell-free form and participation in anti-inflammation in its soluble form, exhibiting cytokine-like functions. We review the biological functions of CD163 which have been discovered until now. It seems apparent from this review that CD163 scavenger receptor can be used as biomarker in different diseases and as a valuable diagnostic parameter for prognosis of many diseases especially inflammatory disorders and sepsis.
Collapse
|
40
|
Soluble CD163 modulates cytokine production by peripheral blood mononuclear cells stimulated by Dermatophagoides pteronyssinus allergens in vitro. Adv Med Sci 2016; 61:249-254. [PMID: 27100336 DOI: 10.1016/j.advms.2016.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 12/24/2022]
Abstract
PURPOSE The CD163 is a scavenger receptor expressed exclusively on monocytes/macrophages which has been shown to exert anti-inflammatory effects. The aim of this study was to evaluate the effect of exogenous sCD163 on production of selected cytokines by peripheral blood mononuclear cells (PBMC) of house dust mite allergic patients (AAPs) stimulated in vitro with Dermatophagoides pteronyssinus (Dp) allergens. PATIENTS AND METHODS The study was performed in 24 AAPs and 12 healthy control subjects (HCs). Peripheral blood mononuclear cells were cultured for up to 144h (T144) in the presence of Dp extract with or without sCD163. Concentration of interleukin (IL) - 10, IL-13 and transforming growth factor beta (TGF-β) was evaluated in the cell culture supernatants using ELISA. Expression of the selected cytokines was evaluated in cell culture lysates using Taqman-based real time polymerase chain reaction (RT-PCR). RESULTS Dp-stimulated PBMC from AAPs released more IL-10 and IL-13 than those from HCs. The greatest up-regulation of IL-10 expression was seen at T6, while that of IL-13 was delayed. Soluble CD163 augmented production of IL-10 in response to Dp stimulation. No significant effect of sCD163 on production of IL-13 and IL-10 by PBMC of HCs could be demonstrated. CONCLUSIONS In AAPs sCD163 modulates the immune response to Dp allergens potentiating anti-inflammatory, homeostatic mechanisms.
Collapse
|
41
|
Gren ST, Janciauskiene S, Sandeep S, Jonigk D, Kvist PH, Gerwien JG, Håkansson K, Grip O. The protease inhibitor cystatin C down-regulates the release of IL-β and TNF-α in lipopolysaccharide activated monocytes. J Leukoc Biol 2016; 100:811-822. [DOI: 10.1189/jlb.5a0415-174r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/04/2016] [Indexed: 01/14/2023] Open
|
42
|
López-Dupla M, Maymó-Masip E, Martínez E, Domingo P, Leal M, Peraire J, Viladés C, Veloso S, Arnedo M, Ferrando-Martínez S, Beltrán-Debón R, Alba V, Gatell JM, Vendrell J, Vidal F, Chacón MR. HIV-1/HAART-Related Lipodystrophy Syndrome (HALS) Is Associated with Decreased Circulating sTWEAK Levels. PLoS One 2015; 10:e0144789. [PMID: 26658801 PMCID: PMC4684375 DOI: 10.1371/journal.pone.0144789] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/22/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Obesity and HIV-1/HAART-associated lipodystrophy syndrome (HALS) share clinical, pathological and mechanistic features. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a multifunctional cytokine that plays an important role in obesity and related diseases. We sought to explore the relationship between HALS and circulating levels of soluble (s) TWEAK and its scavenger receptor sCD163. METHODS This was a cross-sectional multicenter study of 120 HIV-1-infected patients treated with a stable HAART regimen; 56 with overt HALS and 64 without HALS. Epidemiological and clinical variables were determined. Serum levels of sTWEAK and sCD163 levels were measured by ELISA. Results were analyzed with Student's t-test, Mann-Whitney U and χ2 test. Pearson and Spearman correlation were used to estimate the strength of association between variables. RESULTS Circulating sTWEAK was significantly decreased in HALS patients compared with non-HALS patients (2.81±0.2 vs. 2.94±0.28 pg/mL, p = 0.018). No changes were observed in sCD163 levels in the studied cohorts. On multivariate analysis, a lower log sTWEAK concentration was independently associated with the presence of HALS (OR 0.027, 95% CI 0.001-0.521, p = 0.027). CONCLUSIONS HALS is associated with decreased sTWEAK levels.
Collapse
Affiliation(s)
- Miguel López-Dupla
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Elsa Maymó-Masip
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Tarragona, Spain
| | | | - Pere Domingo
- Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Leal
- Hospital Universitario Virgen del Rocio, IBIS, Universidad de Sevilla, Sevilla, Spain
| | - Joaquim Peraire
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Consuelo Viladés
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Sergi Veloso
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Mireia Arnedo
- Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | | | - Raúl Beltrán-Debón
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Verónica Alba
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | | | - Joan Vendrell
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Tarragona, Spain
| | - Francesc Vidal
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Matilde R. Chacón
- Hospital Universitari Joan XXIII. IISPV, Universitat Rovira i Virgili, Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Tarragona, Spain
| |
Collapse
|
43
|
Maras JS, Maiwall R, Harsha HC, Das S, Hussain MS, Kumar C, Bihari C, Rastogi A, Kumar M, Trehanpati N, Sharma S, Pandey A, Sarin SK. Dysregulated iron homeostasis is strongly associated with multiorgan failure and early mortality in acute-on-chronic liver failure. Hepatology 2015; 61:1306-20. [PMID: 25475192 DOI: 10.1002/hep.27636] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 10/22/2014] [Accepted: 11/20/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Acute-on-chronic liver failure (ACLF) is an ailment with high incidence of multiorgan failure (MOF) and consequent mortality. Dysregulated iron homeostasis and macrophage dysfunction are linked to increased incidence of MOF. We investigated whether a panel of circulating iron-regulating proteins are associated with development of MOF and can predict 15- or 30-day mortality in ACLF patients. One hundred twenty patients with ACLF, 20 patients with compensated cirrhosis, and 20 healthy controls were studied. Relative protein expression profiling was performed in the derivative cohort and confirmed in the validation cohort. A panel of iron regulators and indices were determined. Multiparametric flow cytometry for quantitation of labile iron pool (LIP) was performed. Validation studies confirmed lower serum transferrin (Tf) and ceruloplasmin levels in ACLF and ACLF-MOF, compared to patients with cirrhosis and controls (P < 0.01). Serum iron and ferritin levels were markedly elevated (P < 0.001; P < 0.05) and hepcidin levels were lower (P < 0.001) in ACLF patients with MOF than those without and other groups (P < 0.001). Percentage Tf saturation (%SAT) was higher in ACLF-MOF (39.2%; P < 0.001) and correlated with poor outcome (hazard ratio: 6.970; P < 0.01). Intracellular LIP indices were significantly elevated in the subsets of circulating macrophages in ACLF-MOF, compared to other groups (P < 0.01). Whereas expression of iron-regulatory genes was markedly down-regulated, genes related to endoplasmic reticulum stress, apoptosis, and inflammation were up-regulated in ACLF patients, compared to patients with cirrhosis. Severe dysregulation of autophagy mechanisms was also observed in the former. CONCLUSIONS Iron metabolism and transport are severely deranged in ACLF patients and more so in those with MOF. %SAT, circulating hepcidin, and LIP in macrophages correlate with disease severity and %SAT could be used for early prognostication in ACLF patients.
Collapse
Affiliation(s)
- Jaswinder Singh Maras
- Departments of Research, Institute of Liver & Biliary Sciences (ILBS), New Delhi, India
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ambegaokar SS, Kolson DL. Heme oxygenase-1 dysregulation in the brain: implications for HIV-associated neurocognitive disorders. Curr HIV Res 2015; 12:174-88. [PMID: 24862327 PMCID: PMC4155834 DOI: 10.2174/1570162x12666140526122709] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 01/20/2014] [Accepted: 01/27/2014] [Indexed: 12/17/2022]
Abstract
Heme oxygenase-1 (HO-1) is a highly inducible and ubiquitous cellular enzyme that subserves cytoprotective responses to toxic insults, including inflammation and oxidative stress. In neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease and multiple sclerosis, HO-1 expression is increased, presumably reflecting an endogenous neuroprotective response against ongoing cellular injury. In contrast, we have found that in human immunodeficiency virus (HIV) infection of the brain, which is also associated with inflammation, oxidative stress and neurodegeneration, HO-1 expression is decreased, likely reflecting a unique role for HO-1 deficiency in neurodegeneration pathways activated by HIV infection. We have also shown that HO-1 expression is significantly suppressed by HIV replication in cultured macrophages which represent the primary cellular reservoir for HIV in the brain. HO-1 deficiency is associated with release of neurotoxic levels of glutamate from both HIV-infected and immune-activated macrophages; this glutamate-mediated neurotoxicity is suppressed by pharmacological induction of HO-1 expression in the macrophages. Thus, HO-1 induction could be a therapeutic strategy for neuroprotection against HIV infection and other neuroinflammatory brain diseases. Here, we review various stimuli and signaling pathways regulating HO-1 expression in macrophages, which could promote neuronal survival through HO-1-modulation of endogenous antioxidant and immune modulatory pathways, thus limiting the oxidative stress that can promote HIV disease progression in the CNS. The use of pharmacological inducers of endogenous HO-1 expression as potential adjunctive neuroprotective therapeutics in HIV infection is also discussed.
Collapse
Affiliation(s)
| | - Dennis L Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 280 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA 19104, USA.
| |
Collapse
|
45
|
Gerngross L, Lehmicke G, Belkadi A, Fischer T. Role for cFMS in maintaining alternative macrophage polarization in SIV infection: implications for HIV neuropathogenesis. J Neuroinflammation 2015; 12:58. [PMID: 25886134 PMCID: PMC4381451 DOI: 10.1186/s12974-015-0272-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/26/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Macrophage-colony stimulating factor (M-CSF) has been implicated in HIV neuropathogenesis through its ability to modulate activation of macrophages (MΦs) and microglia, as well as enhance the susceptibility of these cells to infection and promote virus production. We have recently reported that MΦs accumulating perivascularly and within nodular lesions in archival brain tissue of simian immunodeficiency virus (SIV)-infected rhesus macaques with encephalitis (SIVE) express M-CSF. In contrast, IL-34, which shares the same receptor, cFMS, was observed more often in parenchymal cells. METHODS Frontal white and grey matter from non-infected and SIV-infected rhesus macaques with and without SIVE were examined by single- and double-label immunohistochemistry for M-CSF, IL-34, and CD163 expression. Primary rhesus macaque and human peripheral blood mononuclear cells were cultured with and without 2.5 ng/ml M-CSF or IL-34 alone and with 470 nM or 4.7 μM of GW2580, a receptor tyrosine kinase inhibitor with high specificity for cFMS. After 24 h, cells were analyzed by flow cytometry to examine the effect of these cytokines on promoting an M2 monocyte/MΦ phenotype. RESULTS Here, we demonstrate that in SIVE brain, accumulating M-CSF(+) MΦs are also CD163(+), while IL-34 does not appear to co-localize significantly with CD163 in the parenchyma. We further demonstrate that M-CSF and IL-34 are expressed by neurons in normal brain but are altered in SIV and SIVE. Through in vitro studies, we show that M-CSF and IL-34 upregulate CD163, a marker for type 2 activation of MΦs (M2), by primary monocytes, which is attenuated by the addition of GW2580. CONCLUSIONS Together, these data suggest that both cFMS ligands may promote and/or prolong M2 activation of MΦs and microglia in brains of SIV-infected animals with encephalitis. As such, cFMS signaling may be an attractive target for eliminating long-lived MΦ reservoirs of HIV infection in brain, as well as other tissues.
Collapse
Affiliation(s)
- Lindsey Gerngross
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, MERB, Room 748, Philadelphia, PA, 19140, USA.
| | - Gabrielle Lehmicke
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, MERB, Room 748, Philadelphia, PA, 19140, USA.
| | - Aghilas Belkadi
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, MERB, Room 748, Philadelphia, PA, 19140, USA.
| | - Tracy Fischer
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, MERB, Room 748, Philadelphia, PA, 19140, USA.
| |
Collapse
|
46
|
Rajasuriar R, Kong YY, Nadarajah R, Abdullah NK, Spelman T, Yuhana MY, Ponampalavanar S, Kamarulzaman A, Lewin SR. The CD14 C-260T single nucleotide polymorphism (SNP) modulates monocyte/macrophage activation in treated HIV-infected individuals. J Transl Med 2015; 13:30. [PMID: 25622527 PMCID: PMC4311493 DOI: 10.1186/s12967-015-0391-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/13/2015] [Indexed: 01/25/2023] Open
Abstract
Background HIV-infected individuals have an increased risk of cardiovascular disease (CVD). T-allele carriers of the CD14 C-260T single-nucleotide polymorphism (SNP) have reported increased expression of the LPS-binding receptor, CD14 and inflammation in the general population. Our aim was to explore the relationship of this SNP with monocyte/macrophage activation and inflammation and its association with sub-clinical atherosclerosis in HIV-infected individuals. Methods Patients with no pre-existing CVD risk factors on suppressive antiretroviral therapy were recruited from University Malaya Medical Centre, Malaysia (n = 84). The CD14 C-260T and TLR4 SNPs, Asp299Gly and Thr399Ile were genotyped and soluble(s) CD14 and sCD163 and high-sensitivity C-reactive protein, hsCRP were measured in plasma. Subclinical atherosclerosis was assessed by measuring carotid intima media thickness (cIMT). The association between CD14 C-260T SNP carriage and cIMT was assessed in a multivariable quantile regression model where a p-value of <0.05 was considered significant. Results We found the CD14 C-260T T-allele in 56% of the cohort and evidence of subclinical atherosclerosis in 27%. TT genotype was associated with higher sCD163 (p = 0.009) but only marginally higher sCD14 (p = 0.209) and no difference in hsCRP (p = 0.296) compared to CC/CT. In multivariable analysis, only Framingham risk score was independently associated with higher cIMT while lower sCD163 was trending towards significance. No association was found in TT-genotype carriers and cIMT measurements. Conclusion The CD14 C-260T SNP was associated with increased monocyte activation but not systemic inflammation or cIMT in this HIV-infected cohort with low CVD risk profile.
Collapse
Affiliation(s)
- Reena Rajasuriar
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia. .,Centre of Excellence for Research in AIDS (CERiA), University of Malaya, 50603, Kuala Lumpur, Malaysia. .,Department of Infectious Diseases, Monash University and Alfred Hospital, 3004, Melbourne, Australia.
| | - Yong Yean Kong
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Reshika Nadarajah
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Noor Kamila Abdullah
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Tim Spelman
- Centre for Population Health, Burnet Institute, 3004, Melbourne, Australia.
| | - Muhamad Yazli Yuhana
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia. .,Faculty of Medicine, University Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia.
| | - Sasheela Ponampalavanar
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, 50603, Kuala Lumpur, Malaysia. .,Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, 50603, Kuala Lumpur, Malaysia. .,Department of Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Sharon R Lewin
- Department of Infectious Diseases, Monash University and Alfred Hospital, 3004, Melbourne, Australia. .,Centre for Biomedical Research, Burnet Institute, 3004, Melbourne, Australia. .,Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, 3010, Australia.
| |
Collapse
|
47
|
Fjeldborg K, Møller HJ, Richelsen B, Pedersen SB. Regulation of CD163 mRNA and soluble CD163 protein in human adipose tissue in vitro. J Mol Endocrinol 2014; 53:227-35. [PMID: 25074267 DOI: 10.1530/jme-14-0089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
CD163-positive macrophages are highly expressed in the human adipose tissue (AT) particularly from obese individuals. However, little is known about the regulation of CD163 mRNA and the protein level of sCD163 in human AT. We aimed to examine the regulation of CD163 and sCD163 in AT. Human s.c. AT samples (n=5) were stimulated with dexamethasone (DEX; 200 nmol/l), lipopolysaccharide (LPS; 100 ng/ml), or DEX+LPS for various time periods up to 24 h. Gene expressions of CD163, ADAM17, IL10, and TNFA (TNF) were measured by RT-PCR. Protein levels of sCD163, IL10, and TNFα (TNF) were measured by ELISA. Furthermore, AT was separated into stromal and adipocyte fraction. We found that CD163 mRNA was strongly expressed in the stromal vascular fraction but hardly detectable in the isolated adipocytes. Incubating whole AT with DEX significantly up-regulated CD163 (P<0.001), whereas incubation with LPS had no effects on CD163 (P>0.05). By contrast, the protein level of sCD163 was not affected by DEX (P>0.05), but LPS significantly increased the level of sCD163 and TNFα (P<0.05). This might be due to the concomitant LPS stimulation of ADAM17, which is known to mediate shedding of the extracellular domains of sCD163 and TNFα. Finally, DEX significantly reduced the LPS-induced TNFα release to the incubation medium but had no effects on sCD163. We conclude that the expression of CD163 and the release of sCD163 are differentially regulated in human AT. Moreover, similar to studies on differentiated blood monocytes, TNFα and sCD163 are concomitantly released in human AT by LPS, which also up-regulate ADAM17.
Collapse
MESH Headings
- ADAM Proteins/genetics
- ADAM17 Protein
- Adipocytes/metabolism
- Adipose Tissue/metabolism
- Antigens, CD/blood
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/blood
- Antigens, Differentiation, Myelomonocytic/genetics
- Biomarkers/metabolism
- Gene Expression Regulation
- Humans
- In Vitro Techniques
- Inflammation/genetics
- Inflammation/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cell Surface/blood
- Receptors, Cell Surface/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Karen Fjeldborg
- Departments of Endocrinology and Internal MedicineMEAClinical BiochemistryAarhus University Hospital, Tage Hansensgade 2, DK-8000 Aarhus C, Denmark
| | - Holger J Møller
- Departments of Endocrinology and Internal MedicineMEAClinical BiochemistryAarhus University Hospital, Tage Hansensgade 2, DK-8000 Aarhus C, Denmark
| | - Bjørn Richelsen
- Departments of Endocrinology and Internal MedicineMEAClinical BiochemistryAarhus University Hospital, Tage Hansensgade 2, DK-8000 Aarhus C, Denmark
| | - Steen B Pedersen
- Departments of Endocrinology and Internal MedicineMEAClinical BiochemistryAarhus University Hospital, Tage Hansensgade 2, DK-8000 Aarhus C, Denmark
| |
Collapse
|
48
|
El Kasmi KC, Pugliese SC, Riddle SR, Poth JM, Anderson AL, Frid MG, Li M, Pullamsetti SS, Savai R, Nagel MA, Fini MA, Graham BB, Tuder RM, Friedman JE, Eltzschig HK, Sokol RJ, Stenmark KR. Adventitial fibroblasts induce a distinct proinflammatory/profibrotic macrophage phenotype in pulmonary hypertension. THE JOURNAL OF IMMUNOLOGY 2014; 193:597-609. [PMID: 24928992 DOI: 10.4049/jimmunol.1303048] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophage accumulation is not only a characteristic hallmark but is also a critical component of pulmonary artery remodeling associated with pulmonary hypertension (PH). However, the cellular and molecular mechanisms that drive vascular macrophage activation and their functional phenotype remain poorly defined. Using multiple levels of in vivo (bovine and rat models of hypoxia-induced PH, together with human tissue samples) and in vitro (primary mouse, rat, and bovine macrophages, human monocytes, and primary human and bovine fibroblasts) approaches, we observed that adventitial fibroblasts derived from hypertensive pulmonary arteries (bovine and human) regulate macrophage activation. These fibroblasts activate macrophages through paracrine IL-6 and STAT3, HIF1, and C/EBPβ signaling to drive expression of genes previously implicated in chronic inflammation, tissue remodeling, and PH. This distinct fibroblast-activated macrophage phenotype was independent of IL-4/IL-13-STAT6 and TLR-MyD88 signaling. We found that genetic STAT3 haplodeficiency in macrophages attenuated macrophage activation, complete STAT3 deficiency increased macrophage activation through compensatory upregulation of STAT1 signaling, and deficiency in C/EBPβ or HIF1 attenuated fibroblast-driven macrophage activation. These findings challenge the current paradigm of IL-4/IL-13-STAT6-mediated alternative macrophage activation as the sole driver of vascular remodeling in PH, and uncover a cross-talk between adventitial fibroblasts and macrophages in which paracrine IL-6-activated STAT3, HIF1α, and C/EBPβ signaling are critical for macrophage activation and polarization. Thus, targeting IL-6 signaling in macrophages by completely inhibiting C/EBPβ or HIF1α or by partially inhibiting STAT3 may hold therapeutic value for treatment of PH and other inflammatory conditions characterized by increased IL-6 and absent IL-4/IL-13 signaling.
Collapse
Affiliation(s)
- Karim C El Kasmi
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045;
| | - Steven C Pugliese
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Suzette R Riddle
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Jens M Poth
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Aimee L Anderson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Maria G Frid
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Min Li
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Soni S Pullamsetti
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, University of Giessen and Marburg Lung Center, German Center for Lung Research, D-61231 Bad Nauheim, Germany
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, University of Giessen and Marburg Lung Center, German Center for Lung Research, D-61231 Bad Nauheim, Germany
| | - Maria A Nagel
- Department of Neurology, University of Colorado Denver, School of Medicine, Aurora, CO 80045
| | - Mehdi A Fini
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Brian B Graham
- Program in Translational Lung Research, Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Rubin M Tuder
- Program in Translational Lung Research, Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Jacob E Friedman
- Division of Biochemistry and Molecular Genetics, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045; and
| | - Holger K Eltzschig
- Department of Anesthesiology, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Ronald J Sokol
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Kurt R Stenmark
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045;
| |
Collapse
|
49
|
Hull TD, Agarwal A, George JF. The mononuclear phagocyte system in homeostasis and disease: a role for heme oxygenase-1. Antioxid Redox Signal 2014; 20:1770-88. [PMID: 24147608 PMCID: PMC3961794 DOI: 10.1089/ars.2013.5673] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/22/2013] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Heme oxygenase-1 (HO-1) is a potential therapeutic target in many diseases, especially those mediated by oxidative stress and inflammation. HO-1 expression appears to regulate the homeostatic activity and distribution of mononuclear phagocytes (MP) in lymphoid tissue under physiological conditions. It also regulates the ability of MP to modulate the inflammatory response to tissue injury. RECENT ADVANCES The induction of HO-1 within MP-particularly macrophages and dendritic cells-modulates the effector functions that they acquire after activation. These effector functions include cytokine production, surface receptor expression, maturation state, and polarization toward a pro- or anti-inflammatory phenotype. The importance of HO-1 in MP is emphasized by their expression of specific receptors that primarily function to ingest heme-containing substrate and deliver it to HO-1. CRITICAL ISSUES MP are the first immunological responders to tissue damage. They critically affect the outcome of injury to many organ systems, yet few therapies are currently available to specifically target MP during disease pathogenesis. Elucidation of the role of HO-1 expression in MP may help to direct broadly applicable therapies to clinical use that are based on the immunomodulatory capabilities of HO-1. FUTURE DIRECTIONS Unraveling the complexities of HO-1 expression specifically within MP will more completely define how HO-1 provides cytoprotection in vivo. The use of models in which HO-1 expression is specifically modulated in bone marrow-derived cells will allow for a more complete characterization of its immunoregulatory properties.
Collapse
Affiliation(s)
- Travis D. Hull
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - James F. George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
50
|
Pey P, Pearce RKB, Kalaitzakis ME, Griffin WST, Gentleman SM. Phenotypic profile of alternative activation marker CD163 is different in Alzheimer's and Parkinson's disease. Acta Neuropathol Commun 2014; 2:21. [PMID: 24528486 PMCID: PMC3940003 DOI: 10.1186/2051-5960-2-21] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/27/2014] [Indexed: 01/07/2023] Open
Abstract
Background Microglial activation is a pathological feature common to both Alzheimer’s and Parkinson’s diseases (AD and PD). The classical activation involves release of pro-inflammatory cytokines and reactive oxygen species. This is necessary for maintenance of tissue homeostasis and host defense, but can cause bystander damage when the activation is sustained and uncontrolled. In recent years the heterogeneous nature of microglial activation states in neurodegenerative diseases has become clear and the focus has shifted to alternative activation states that promote tissue maintenance and repair. We studied the distribution of CD163, a membrane-bound scavenger receptor found on perivascular macrophages. CD163 has an immunoregulatory function, and has been found in the parenchyma in other inflammatory diseases e.g. HIV-encephalitis and multiple sclerosis. In this study, we used immunohistochemistry to compare CD163 immunoreactivity in 31 AD cases, 27 PD cases, and 16 control cases. Associations of microglia with pathological hallmarks of AD and PD were investigated using double immunofluorescence. Results Parenchymal microglia were found to be immunoreactive for CD163 in all of the AD cases, and to a lesser extent in PD cases. There was prominent staining of CD163 immunoreactive microglia in the frontal and occipital cortices of AD cases, and in the brainstem of PD cases. Many of them were associated with Aß plaques in both diseases, and double staining with CD68 demonstrates their phagocytic capability. Leakage of fibrinogen was observed around compromised blood vessels, raising the possibility these microglia might have originated from the periphery. Conclusions Increase in microglia’s CD163 immunoreactivity was more significant in AD than PD, and association of CD163 immunoreactive microglia with Aβ plaques indicate microglia’s attraction towards extracellular protein pathology, i.e. extracellular aggregates of Aβ as compared to intracellular Lewy Bodies in PD. Double staining with CD163 and CD68 might point towards their natural inclination to phagocytose plaques. Fibrinogen leakage and compromise of the blood brain barrier raise the possibility that these are not resident microglia, but systemic macrophages infiltrating the brain.
Collapse
|