1
|
Ahrazoglu T, Kluczny JI, Kleimann P, Irschfeld LM, Nienhaus FT, Bönner F, Gerdes N, Temme S. Design of a Robust Flow Cytometric Approach for Phenotypical and Functional Analysis of Human Monocyte Subsets in Health and Disease. Biomolecules 2024; 14:1251. [PMID: 39456184 PMCID: PMC11506830 DOI: 10.3390/biom14101251] [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: 08/02/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Human monocytes can be subdivided into phenotypically and functionally different classical, intermediate and non-classical monocytes according to the cell surface expression of CD14 and CD16. A precise identification and characterisation of monocyte subsets is necessary to unravel their role in inflammatory diseases. Here, we compared three different flow cytometric strategies (A-C) and found that strategy C, which included staining against CD11b, HLA-DR, CD14 and CD16, followed by several gating steps, most reliably identified monocyte subtypes in blood samples from healthy volunteers and from patients with stable coronary heart disease (CHD) or ST-elevation myocardial infarction (STEMI). Additionally, we established a fixation and permeabilisation protocol to enable the analysis of intracellular markers. We investigated the phagocytosis of lipid nanoparticles, the uptake of 2-NBD-glucose and the intracellular levels of CD74 and HLA-DM. This revealed that classical and intermediate monocytes from patients with STEMI showed the highest uptake of 2-NBD-glucose, whereas classical and intermediate monocytes from patients with CHD took up the largest amounts of lipid nanoparticles. Interestingly, intermediate monocytes had the highest expression level of HLA-DM. Taken together, we present a robust flow cytometric approach for the identification and functional characterisation of monocyte subtypes in healthy humans and patients with diseases.
Collapse
Affiliation(s)
- Talia Ahrazoglu
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (T.A.); (J.I.K.)
| | - Jennifer Isabel Kluczny
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (T.A.); (J.I.K.)
| | - Patricia Kleimann
- Institute of Molecular Cardiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Lisa-Marie Irschfeld
- Department of Radiation Oncology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Fabian Theodor Nienhaus
- Department of Cardiology, Pulmonology and Vascular Medicine, Faculty of Medicine, University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (F.T.N.); (F.B.); (N.G.)
| | - Florian Bönner
- Department of Cardiology, Pulmonology and Vascular Medicine, Faculty of Medicine, University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (F.T.N.); (F.B.); (N.G.)
| | - Norbert Gerdes
- Department of Cardiology, Pulmonology and Vascular Medicine, Faculty of Medicine, University Hospital, Heinrich-Heine University, 40225 Düsseldorf, Germany; (F.T.N.); (F.B.); (N.G.)
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Sebastian Temme
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (T.A.); (J.I.K.)
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany
| |
Collapse
|
2
|
Monif M, Sequeira RP, Muscat A, Stuckey S, Sanfilippo PG, Minh V, Loftus N, Voo V, Fazzolari K, Moss M, Maltby VE, Nguyen AL, Wesselingh R, Seery N, Nesbitt C, Baker J, Dwyer C, Taylor L, Rath L, Van der Walt A, Marriott M, Kalincik T, Lechner-Scott J, O'Brien TJ, Butzkueven H. CLADIN- CLADribine and INnate immune response in multiple sclerosis - A phase IV prospective study. Clin Immunol 2024; 265:110304. [PMID: 38964633 DOI: 10.1016/j.clim.2024.110304] [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: 04/26/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Cladribine (Mavenclad®) is an oral treatment for relapsing remitting MS (RRMS), but its mechanism of action and its effects on innate immune responses in unknown. This study is a prospective Phase IV study of 41 patients with RRMS, and aims to investigate the mechanism of action of cladribine on peripheral monocytes, and its impact on the P2X7 receptor. There was a significant reduction in monocyte count in vivo at week 1 post cladribine administration, and the subset of cells being most impacted were the CD14lo CD16+ 'non-classical' monocytes. Of the 14 cytokines measured in serum, CCL2 levels increased at week 1. In vitro, cladrabine induced a reduction in P2X7R pore as well as channel activity. This study demonstrates a novel mechanism of action for cladribine. It calls for studying potential benefits of cladribine in progressive forms of MS and other neurodegenerative diseases where innate immune related inflammation is implicated in disease pathogenesis.
Collapse
Affiliation(s)
- Mastura Monif
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Neurology, Melbourne Health, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, Australia; Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.
| | - Richard P Sequeira
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Andrea Muscat
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Sian Stuckey
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Paul G Sanfilippo
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Viet Minh
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia; School of Nursing, Midwifery and Paramedicine, Australian Catholic University, Melbourne, VIC, Australia
| | - Naomi Loftus
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Veronica Voo
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | | | - Melinda Moss
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Vicki E Maltby
- John Hunter Hospital, Department of Neurology, New Lambton Heights, NSW, Australia; School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Ai-Lan Nguyen
- Department of Neurology, Melbourne Health, Melbourne, VIC, Australia; Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Robb Wesselingh
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Nabil Seery
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Cassie Nesbitt
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia; Department of Neurology, Barwon Health, Melbourne, VIC, Australia
| | - Josephine Baker
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Chris Dwyer
- Department of Neurology, Melbourne Health, Melbourne, VIC, Australia
| | - Lisa Taylor
- Department of Neurology, Melbourne Health, Melbourne, VIC, Australia
| | - Louise Rath
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Anneke Van der Walt
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Mark Marriott
- Department of Neurology, Melbourne Health, Melbourne, VIC, Australia; Department of Medicine, University of Melbourne, Melbourne, VIC, Australia; Department of Neurology, Eastern Health, Melbourne, VIC, Australia
| | - Tomas Kalincik
- Department of Neurology, Melbourne Health, Melbourne, VIC, Australia; Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Jeannette Lechner-Scott
- John Hunter Hospital, Department of Neurology, New Lambton Heights, NSW, Australia; School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Hays KE, Pfaffinger JM, Ryznar R. The interplay between gut microbiota, short-chain fatty acids, and implications for host health and disease. Gut Microbes 2024; 16:2393270. [PMID: 39284033 PMCID: PMC11407412 DOI: 10.1080/19490976.2024.2393270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Short-chain fatty acids (SCFAs) - acetate, propionate, and butyrate - are important bacterial fermentation metabolites regulating many important aspects of human physiology. Decreases in the concentrations of any or multiple SCFAs are associated with various detrimental effects to the host. Previous research has broadly focused on gut microbiome produced SCFAs as a group, with minimal distinction between acetate, propionate, and butyrate independently, each with significantly different host effects. In this review, we comprehensively delineate the roles of these SCFAs with emphasis on receptor affinity, signaling pathway involvement, and net host physiologic effects. Butyrate is highlighted due to its unique role in gastrointestinal-associated functions, especially maintaining gut barrier integrity. Butyrate functions by promoting epithelial tight junctions, serving as fuel for colonocyte ATP production, and modulating the immune system. Interaction with the immune system occurs locally in the gastrointestinal tract and systemically in the brain. Investigation into research conducted on butyrate production pathways and specific bacterial players involved highlights a unique risk associated with use of gram-positive targeted antibiotics. We review and discuss evidence showing the relationship between the butyrate-producing gram-positive genus, Roseburia, and susceptibility to commonly prescribed, widely used gram-positive antibiotics. Considering gut microbiome implications when choosing antibiotic therapy may benefit health outcomes in patients.
Collapse
Affiliation(s)
- Kallie E Hays
- Doctor of Osteopathic Medicine Program, Rocky Vista University College of Osteopathic Medicine, Englewood, CO, USA
| | - Jacob M Pfaffinger
- Doctor of Osteopathic Medicine Program, Rocky Vista University College of Osteopathic Medicine, Englewood, CO, USA
| | - Rebecca Ryznar
- Department of Biomedical Sciences, Rocky Vista University College of Osteopathic Medicine, Englewood, CO, USA
| |
Collapse
|
4
|
Ludwig K, Chichelnitskiy E, Kühne JF, Wiegmann B, Iske J, Ledwoch N, Ius F, Beushausen K, Keil J, Iordanidis S, Rojas SV, Salman J, Knoefel AK, Haverich A, Warnecke G, Falk CS. CD14 highCD16 + monocytes are the main producers of Interleukin-10 following clinical heart transplantation. Front Immunol 2023; 14:1257526. [PMID: 37936714 PMCID: PMC10627027 DOI: 10.3389/fimmu.2023.1257526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Following heart transplantation, a cascade of immunological responses is initiated influencing the clinical outcome and long-term survival of the transplanted patients. The anti-inflammatory cytokine interleukin-10 (IL-10) was shown to be elevated in the blood of heart transplant recipients directly after transplantation but the releasing cell populations and the composition of lymphocyte subsets following transplantation have not been thoroughly studied. Methods We identified immune cells by immunophenotyping and analyzed intracellular IL-10 production in peripheral blood mononuclear cells (PBMC) of heart transplanted patients (n= 17) before, directly after and 24h post heart transplantation. The cells were stimulated with lipopolysaccharide or PMA/Ionomycin to enhance cytokine production within leukocytes in vitro. Results and discussion We demonstrate that intermediate monocytes (CD14highCD16+), but not CD8+ T cells, CD4+ T cells, CD56+ NK cells or CD20+ B cells appeared to be the major IL-10 producers within patients PBMC following heart transplantation. Consequently, the absolute monocyte count and the ratio of intermediate monocytes to classical monocytes (CD14+CD16-) were specifically increased in comparison to pre transplant levels. Hence, this population of monocytes, which has not been in the focus of heart transplantation so far, may be an important modulator of clinical outcome and long-term survival of heart transplant recipients. Alteration of blood-circulating monocytes towards a CD14highCD16+ phenotype could therefore shift the pro-inflammatory immune response towards induction of graft tolerance, and may pave the way for the optimization of immunosuppression.
Collapse
Affiliation(s)
- Kristina Ludwig
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
- Department of Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Jenny F. Kühne
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Bettina Wiegmann
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Hannover Medical School, Hannover, Germany
- DZL, German Center for Lung Diseases, BREATH site, Hannover, Germany
| | - Jasper Iske
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
| | - Nadine Ledwoch
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Fabio Ius
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- DZL, German Center for Lung Diseases, BREATH site, Hannover, Germany
| | - Kerstin Beushausen
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Jana Keil
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Susanne Iordanidis
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Sebastian V. Rojas
- Heart and Diabetes Center Nordrhein-Westfalen, University Hospital Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Jawad Salman
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Ann-Kathrin Knoefel
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Gregor Warnecke
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Christine S. Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
- DZL, German Center for Lung Diseases, BREATH site, Hannover, Germany
- DZIF, German Center for Infection Research, TTU-IICH, Hannover, Germany
| |
Collapse
|
5
|
Cossarizza A, Cozzi-Lepri A, Mattioli M, Paolini A, Neroni A, De Biasi S, Tartaro DL, Borella R, Fidanza L, Gibellini L, Beghetto B, Roncaglia E, Nardini G, Milic J, Menozzi M, Cuomo G, Digaetano M, Orlando G, Borghi V, Guaraldi G, Mussini C. Evaluating immunological and inflammatory changes of treatment-experienced people living with HIV switching from first-line triple cART regimens to DTG/3TC vs. B/F/TAF: the DEBATE trial. Front Immunol 2023; 14:1279390. [PMID: 37908359 PMCID: PMC10613634 DOI: 10.3389/fimmu.2023.1279390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/21/2023] [Indexed: 11/02/2023] Open
Abstract
Background The aim of this randomized clinical trial (RCT) was to compare immunological changes in virally suppressed people living with HIV (PLWH) switching from a three-drug regimen (3DR) to a two-drug regimen (2DR). Methods An open-label, prospective RCT enrolling PLWH receiving a 3DR who switched to bictegravir/emtricitabine/tenofovir alafenamide (B/F/TAF) or dolutegravir/lamivudine (DTG/3TC) was performed. Blood was taken at baseline and months 6 and 12. The primary outcome was the change in CD4+ or CD8+ T-cell counts and CD4/CD8 ratio over time points. The secondary outcomes were the changes in immunological and inflammatory parameters. Parametric mixed-linear models with random intercepts and slopes were fitted separately for each marker after controlling for potential confounders. Results Between the two arms (33 PLWH each), there was no difference in CD4+ or CD8+ T cells, CD4/CD8 ratio, and IL-6 trajectories. PLWH switching to DTG/3TC had increased levels of both transitional memory and terminally differentiated CD4+ T cells (arm-time interaction p-value = 0.02) and to a lesser extent for the corresponding CD8+ T-cell subsets (p = 0.09). Significantly lower levels of non-classical monocytes were detected in the B/F/TAF arm at T6 (diff = -6.7 cells/mm3; 95% CI; -16, +2.6; p-value for interaction between arm and time = 0.03). All differences were attenuated at T12. Conclusion No evidence for a difference in absolute CD4+ and CD8+ T-cell counts, CD4/CD8 ratio, and IL-6 trajectories by study arm over 12 months was found. PLWH on DTG/3TC showed higher levels of terminally differentiated and exhausted CD4+ and CD8+ T lymphocytes and non-classical monocytes at T6. Further studies are warranted to better understand the clinical impact of our results. Clinical Trial Registration https://clinicaltrials.gov, identifier NCT04054089.
Collapse
Affiliation(s)
- Andrea Cossarizza
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Alessandro Cozzi-Lepri
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation (CREME), Institute for Global Health, University College London (UCL), London, United Kingdom
| | - Marco Mattioli
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Annamaria Paolini
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Anita Neroni
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Sara De Biasi
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Domenico Lo Tartaro
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Lucia Fidanza
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Lara Gibellini
- Chair of Pathology and Immunology, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Barbara Beghetto
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrica Roncaglia
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Nardini
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marianna Menozzi
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Gianluca Cuomo
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Margherita Digaetano
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Gabriella Orlando
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Vanni Borghi
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Giovanni Guaraldi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Cristina Mussini
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| |
Collapse
|
6
|
O’Connor KW, Liu T, Kim S, Briseño CG, Georgopoulos K, Murphy TL, Murphy KM. Bcl6, Irf2, and Notch2 promote nonclassical monocyte development. Proc Natl Acad Sci U S A 2023; 120:e2220853120. [PMID: 37607223 PMCID: PMC10469339 DOI: 10.1073/pnas.2220853120] [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: 12/07/2022] [Accepted: 07/28/2023] [Indexed: 08/24/2023] Open
Abstract
Ly6Clo monocytes are a myeloid subset that specializes in the surveillance of vascular endothelium. Ly6Clo monocytes have been shown to derive from Ly6Chi monocytes. NOTCH2 signaling has been implicated as a trigger for Ly6Clo monocyte development, but the basis for this effect is unclear. Here, we examined the impact of NOTCH2 signaling of myeloid progenitors on the development of Ly6Clo monocytes in vitro. NOTCH2 signaling induced by delta-like ligand 1 (DLL1) efficiently induced the transition of Ly6Chi TREML4- monocytes into Ly6Clo TREML4+ monocytes. We further identified two additional transcriptional requirements for development of Ly6Clo monocytes. Deletion of BCL6 from myeloid progenitors abrogated development of Ly6Clo monocytes. IRF2 was also required for Ly6Clo monocyte development in a cell-intrinsic manner. DLL1-induced in vitro transition into Ly6Clo TREML4+ monocytes required IRF2 but unexpectedly could occur in the absence of NUR77 or BCL6. These results imply a transcriptional hierarchy for these factors in controlling Ly6Clo monocyte development.
Collapse
Affiliation(s)
- Kevin W. O’Connor
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO63110
| | - Tiantian Liu
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO63110
| | - Sunkyung Kim
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO63110
| | - Carlos G. Briseño
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO63110
| | - Katia Georgopoulos
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
| | - Theresa L. Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO63110
| | - Kenneth M. Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO63110
| |
Collapse
|
7
|
Williams H, Mack C, Baraz R, Marimuthu R, Naralashetty S, Li S, Medbury H. Monocyte Differentiation and Heterogeneity: Inter-Subset and Interindividual Differences. Int J Mol Sci 2023; 24:ijms24108757. [PMID: 37240103 DOI: 10.3390/ijms24108757] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The three subsets of human monocytes, classical, intermediate, and nonclassical, show phenotypic heterogeneity, particularly in their expression of CD14 and CD16. This has enabled researchers to delve into the functions of each subset in the steady state as well as in disease. Studies have revealed that monocyte heterogeneity is multi-dimensional. In addition, that their phenotype and function differ between subsets is well established. However, it is becoming evident that heterogeneity also exists within each subset, between health and disease (current or past) states, and even between individuals. This realisation casts long shadows, impacting how we identify and classify the subsets, the functions we assign to them, and how they are examined for alterations in disease. Perhaps the most fascinating is evidence that, even in relative health, interindividual differences in monocyte subsets exist. It is proposed that the individual's microenvironment could cause long-lasting or irreversible changes to monocyte precursors that echo to monocytes and through to their derived macrophages. Here, we will discuss the types of heterogeneity recognised in monocytes, the implications of these for monocyte research, and most importantly, the relevance of this heterogeneity for health and disease.
Collapse
Affiliation(s)
- Helen Williams
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Corinne Mack
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Rana Baraz
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Rekha Marimuthu
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Sravanthi Naralashetty
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Stephen Li
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Chemical Pathology, NSW Health Pathology, Westmead Hospital and Institute of Clinical Pathology and Medical Research, Westmead, NSW 2145, Australia
- . Blacktown/Mt Druitt Clinical School, Blacktown Hospital, Western Sydney University, Blacktown, NSW 2148, Australia
| | - Heather Medbury
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Westmead, NSW 2145, Australia
| |
Collapse
|
8
|
Sangani RG, Deepak V, Anwar J, Patel Z, Ghio AJ. Cigarette Smoking, and Blood Monocyte Count Correlate with Chronic Lung Injuries and Mortality. Int J Chron Obstruct Pulmon Dis 2023; 18:431-446. [PMID: 37034898 PMCID: PMC10076620 DOI: 10.2147/copd.s397667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/27/2023] [Indexed: 04/04/2023] Open
Abstract
Background Cigarette smoking (CS)-related monocytosis contributes to the development of chronic lung injuries via complex mechanisms. We aim to determine correlations between measures of CS and monocytes, their capacities to predict chronic lung diseases, and their associations with mortality. Methods A single-center retrospective study of patients undergoing surgical resection for suspected lung nodules/masses was performed. CS was quantified as cigarettes smoked per day (CPD), duration of smoking, composite pack years (CPY), current smoking status, and smoking cessation years. A multivariate logistic regression analysis was performed. Results Of 382 eligible patients, 88% were ever smokers. In this group, 45% were current smokers with mean CPD of 27.2±40.0. CPY and duration of smoking showed positive linear correlations with percentage monocyte count. Physiologically, CPY was associated with progressive obstruction, hyperinflation, and reduced diffusion capacity (DLCO). Across the quartiles of smoking, there was an accumulation of radiologic and histologic abnormalities. Anthracosis and emphysema were associated with CPD, while lung cancer, respiratory bronchiolitis (RB), emphysema, and honeycombing were statistically related to duration of smoking. Analysis using consecutive CPY showed associations with lung cancer (≥10 and <30), fibrosis (≥20 and <40), RB (≥50), anthracosis and emphysema (≥10 and onwards). Percentage monocytes correlated with organizing pneumonia (OP), fibrosis, and emphysema. The greater CPY increased mortality across the groups. Significant predictors of mortality included percentage monocyte, anemia, GERD, and reduced DLCO. Conclusion Indices of CS and greater monocyte numbers were associated with endpoints of chronic lung disease suggesting a participation in pathogenesis. Application of these easily available metrics may support a chronology of CS-induced chronic lung injuries. While a relative lesser amount of smoking can be associated with lung cancer and fibrosis, greater CPY increases the risk for emphysema. Monocytosis predicted lung fibrosis and mortality. Duration of smoking may serve as a better marker of monocytosis and associated chronic lung diseases.
Collapse
Affiliation(s)
- Rahul G Sangani
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, West Virginia University, Morgantown, WV, USA
- Correspondence: Rahul G Sangani, Section of Pulmonary, Critical Care, and Sleep Medicine, West Virginia University School of Medicine, 1 Medical Center Dr, PO BOX 9166, Morgantown, WV, 26506, USA, Tel +1 304 293-4661 option #2, Fax +1 304-293-3724, Email
| | - Vishal Deepak
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - Javeria Anwar
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - Zalak Patel
- Department of Radiology, West Virginia University, Morgantown, WV, USA
| | | |
Collapse
|
9
|
González-Cuadrado C, Caro-Espada PJ, Chivite-Lacaba M, Utrero-Rico A, Lozano-Yuste C, Gutierrez-Solis E, Morales E, Sandino-Pérez J, Gil-Etayo FJ, Allende-Martínez L, Laguna-Goya R, Paz-Artal E. Hemodialysis-Associated Immune Dysregulation in SARS-CoV-2-Infected End-Stage Renal Disease Patients. Int J Mol Sci 2023; 24:ijms24021712. [PMID: 36675231 PMCID: PMC9865754 DOI: 10.3390/ijms24021712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Patients on hemodialysis show dysregulated immunity, basal hyperinflammation and a marked vulnerability to COVID-19. We evaluated the immune profile in COVID-19 hemodialysis patients and the changes associated with clinical deterioration after the hemodialysis session. Recruited patients included eight hemodialysis subjects with active, PCR-confirmed SARS-CoV-2 infection, five uninfected hemodialysis patients and five healthy controls. In SARS-CoV-2-infected hemodialysis patients TNF-α, IL-6 and IL-8 were particularly increased. Lymphopenia was mostly due to reduction in CD4+ T, B and central memory CD8+ T cells. There was a predominance of classical and intermediate monocytes with reduced HLA-DR expression and enhanced production of pro-inflammatory molecules. Immune parameters were analysed pre- and post-hemodialysis in three patients with COVID-19 symptoms worsening after the hemodialysis session. There was a higher than 2.5-fold increase in GM-CSF, IFN-γ, IL-1β, IL-2, IL-6, IL-17A and IL-21 in serum, and augmentation of monocytes-derived TNF-α, IL-1β and IL-8 and CXCL10 (p < 0.05). In conclusion, COVID-19 in hemodialysis patients associates with alteration of lymphocyte subsets, increasing of pro-inflammatory cytokines and monocyte activation. The observed worsening during the hemodialysis session in some patients was accompanied by augmentation of particular inflammatory cytokines, which might suggest biomarkers and therapeutic targets to prevent or mitigate the hemodialysis-related deterioration during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Cecilia González-Cuadrado
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- Correspondence: (C.G.-C.); (E.P.-A.); Tel.: +34-628-502-629 (C.G.-C.)
| | | | - Marta Chivite-Lacaba
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Alberto Utrero-Rico
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Claudia Lozano-Yuste
- Department of Nephrology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | | | - Enrique Morales
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Nephrology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Department of Medicine, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Justo Sandino-Pérez
- Department of Nephrology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Francisco Javier Gil-Etayo
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, 28009 Madrid, Spain
| | - Luis Allende-Martínez
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, 28009 Madrid, Spain
| | - Rocio Laguna-Goya
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, 28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, 28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Correspondence: (C.G.-C.); (E.P.-A.); Tel.: +34-628-502-629 (C.G.-C.)
| |
Collapse
|
10
|
Zong L, Yang F, Liu S, Gao Y, Xia F, Zheng M, Xu Y. CD8 + T cells mediate antiviral response in severe fever with thrombocytopenia syndrome. FASEB J 2023; 37:e22722. [PMID: 36571509 DOI: 10.1096/fj.202201343rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/27/2022]
Abstract
Severe fever with thrombocytopenia syndrome (SFTS), which is caused by a novel Bunyavirus, has gradually become a threatening infectious disease in rural areas of Asia. Studies have identified a severe cytokine storm and impaired humoral immune response in SFTS. However, the cellular immune response to SFTS virus (SFTSV) infection remains largely unknown. Here we report that SFTS patients had a cytokine storm accompanied by high levels of chemokines. CD8+ T cells in peripheral blood mononuclear cells of SFTS patients exhibited a more activated phenotype and enhanced the antiviral responses. They increased the expression of CD69 and CD25, secreted a higher level of IFN-γ and granzyme, and had a stronger proliferative ability than in healthy controls. In convalescent SFTS patients, the expression of CD69 and CD25 on CD8+ T cells was reduced. In addition, we found the ratio and cellularity of CD14+ CD16+ intermediate monocytes were increased in peripheral blood of SFTS patients. Both the expression of C-X-C motif chemokine ligand 10 (CXCL10) on CD14+ CD16+ intermediate monocytes and the expression of C-X-C motif chemokine receptor 3 (CXCR3) on CD8+ T cells increased dramatically in SFTS patients. Our studies reveal a potential pathway that CD8+ T cells rapidly activate and are mostly recruited by intermediate monocytes through CXCL10 in SFTSV infection. Our results may be of clinical relevance for further treatment and discharge instructions in SFTSV infections.
Collapse
Affiliation(s)
- Lu Zong
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Yang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Siyu Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yufeng Gao
- Department of Infectious Diseases, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fang Xia
- Department of Clinical Laboratory, People's Hospital of Hanshan County, Maanshan, China
| | - Meijuan Zheng
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Głuchowska A, Cysewski D, Baj-Krzyworzeka M, Szatanek R, Węglarczyk K, Podszywałow-Bartnicka P, Sunderland P, Kozłowska E, Śliwińska MA, Dąbrowski M, Sikora E, Mosieniak G. Unbiased proteomic analysis of extracellular vesicles secreted by senescent human vascular smooth muscle cells reveals their ability to modulate immune cell functions. GeroScience 2022; 44:2863-2884. [PMID: 35900662 PMCID: PMC9768090 DOI: 10.1007/s11357-022-00625-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/12/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis, a common age-related disease, is characterized by intense immunological activity. Atherosclerotic plaque is composed of endothelial cells, vascular smooth muscle cells (VSMCs), lipids and immune cells infiltrating from the blood. During progression of the disease, VSMCs undergo senescence within the plaque and secrete SASP (senescence-associated secretory phenotype) factors that can actively modulate plaque microenvironment. We demonstrated that senescent VSMCs secrete increased number of extracellular vesicles (senEVs). Based on unbiased proteomic analysis of VMSC-derived EVs and of the soluble fraction of SASP (sSASP), more than 900 proteins were identified in each of SASP compartments. Comparison of the composition of VMSC-derived EVs with the SASP atlas revealed several proteins, including Serpin Family F Member 1 (SERPINF1) and Thrombospondin 1 (THBS1), as commonly upregulated components of EVs secreted by senescent VSMCs and fibroblasts. Among soluble SASP factors, only Growth Differentiation Factor 15 (GDF15) was universally increased in the secretome of senescent VSMCs, fibroblasts, and epithelial cells. Bioinformatics analysis of EV proteins distinguished functionally organized protein networks involved in immune cell function regulation. Accordingly, EVs released by senescent VSMCs induced secretion of IL-17, INFγ, and IL-10 by T cells and of TNFα produced by monocytes. Moreover senEVs influenced differentiation of monocytes favoring mix M1/M2 polarization with proinflammatory characteristics. Altogether, our studies provide a complex, unbiased analysis of VSMC SASP and prove that EVs derived from senescent VSMCs influence the cytokine milieu by modulating immune cell activity. Our results strengthen the role of senescent cells as an important inducer of inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Agata Głuchowska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Clinical Research Centre, Medical University of Bialystok, Białystok, Poland
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Rafał Szatanek
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | | | - Piotr Sunderland
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Ewa Kozłowska
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology University of Warsaw, Warsaw, Poland
| | - Małgorzata A Śliwińska
- Laboratory of Imaging Tissue Structure and Function, Nencki Institute of Experimental Biology Polish Academy of Sciences, Warsaw, Poland
| | - Michał Dąbrowski
- Laboratory of Bioinformatics, Neurobiology Centre, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Grażyna Mosieniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland.
| |
Collapse
|
12
|
Patysheva M, Frolova A, Larionova I, Afanas'ev S, Tarasova A, Cherdyntseva N, Kzhyshkowska J. Monocyte programming by cancer therapy. Front Immunol 2022; 13:994319. [PMID: 36341366 PMCID: PMC9631446 DOI: 10.3389/fimmu.2022.994319] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/27/2022] [Indexed: 08/27/2023] Open
Abstract
Monocytes in peripheral blood circulation are the precursor of essential cells that control tumor progression, that include tumor-associated macrophages (TAMs), dendritic cells (DCs) and myeloid-derive suppressor cells (MDSC). Monocytes-derived cells orchestrate immune reactions in tumor microenvironment that control disease outcome and efficiency of cancer therapy. Four major types of anti-cancer therapy, surgery, radiotherapy, chemotherapy, and most recent immunotherapy, affect tumor-associated macrophage (TAM) polarization and functions. TAMs can also decrease the efficiency of therapy in a tumor-specific way. Monocytes is a major source of TAMs, and are recruited to tumor mass from the blood circulation. However, the mechanisms of monocyte programming in circulation by different therapeutic onsets are only emerging. In our review, we present the state-of-the art about the effects of anti-cancer therapy on monocyte progenitors and their dedifferentiation, on the content of monocyte subpopulations and their transcriptional programs in the circulation, on their recruitment into tumor mass and their potential to give origin for TAMs in tumor-specific microenvironment. We have also summarized very limited available knowledge about genetics that can affect monocyte interaction with cancer therapy, and highlighted the perspectives for the therapeutic targeting of circulating monocytes in cancer patients. We summarized the knowledge about the mediators that affect monocytes fate in all four types of therapies, and we highlighted the perspectives for targeting monocytes to develop combined and minimally invasive anti-cancer therapeutic approaches.
Collapse
Affiliation(s)
- Marina Patysheva
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Laboratory of Tumor Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Anastasia Frolova
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Laboratory of Tumor Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Laboratory of Genetic Technologies, Siberian State Medical University, Tomsk, Russia
| | - Sergey Afanas'ev
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Department of Abdominal Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Anna Tarasova
- Department of Abdominal Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Nadezhda Cherdyntseva
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Laboratory of Genetic Technologies, Siberian State Medical University, Tomsk, Russia
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Laboratory of Genetic Technologies, Siberian State Medical University, Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany
| |
Collapse
|
13
|
Leishmania infantum Infection of Primary Human Myeloid Cells. Microorganisms 2022; 10:microorganisms10061243. [PMID: 35744760 PMCID: PMC9230042 DOI: 10.3390/microorganisms10061243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 02/04/2023] Open
Abstract
Circulating phagocytic cells often serve as cellular targets for a large number of pathogens such as Leishmania parasites. Studying primary human cells in an infectious context requires lengthy procedures for cell isolation that may affect the analysis performed. Using whole blood and a no-lyse and no-wash flow cytometric assay (NoNo assay), we monitored the Leishmania infantum infection of primary human cells. We demonstrated, using fluorescent parasites, that among monocyte cell populations, L. infantum preferentially infects classical (CD14+CD16−) and intermediate (CD14+CD16+) primary human monocytes in whole blood. Because classical monocytes are the preponderant population, they represent the larger L. infantum reservoir. Moreover, we also found that, concomitantly to monocyte infection, a subset of PMNs is infected early in whole blood. Of interest, in whole blood, PMNs are less infected compared to classical monocytes. Overall, by using this NoNo assay, we provided a novel avenue in our understanding of host–leishmania interactions.
Collapse
|
14
|
Scherberich JE, Fünfstück R, Naber KG. Urinary tract infections in patients with renal insufficiency and dialysis - epidemiology, pathogenesis, clinical symptoms, diagnosis and treatment. GMS INFECTIOUS DISEASES 2021; 9:Doc07. [PMID: 35106269 PMCID: PMC8777485 DOI: 10.3205/id000076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Epidemiological studies show an increasing number of patients worldwide suffering from chronic kidney diseases (CKD), which are associated with a risk for progression to end-stage kidney disease (ESKD). CKD patients stage 2-5, patients with regular chronic dialysis treatment (hemo- or peritoneal dialysis), and patients suffering from kidney allograft dysfunction are at high risk to develop infections, e.g. urinary tract infections (UTI) and/or sepsis (urosepsis). These groups show metabolic disturbance, chronic inflammation, and impaired immunocompetence. Escherichia coli is still the most common pathogen in UTI. A wide variety of other pathogens may be involved in UTI. Urological interventions, catheterization, as well as repeated courses of antibiotics contribute to an increased challenge of antimicrobial resistance. The diagnosis of UTI in CKD is based on standard clinical and laboratory criteria. Pyuria (≥10 leucocytes/µl) is more often observed in patients with oligoanuria and low bacterial colony counts. The treatment strategies for this population are based on the same principles as in patients with normal renal function. However, drugs cleared by the kidney or by dialysis membranes need dose adjustment. Antimicrobials with potential systemic toxicity and nephrotoxicity should be administered with caution.
Collapse
Affiliation(s)
| | | | - Kurt G. Naber
- Department of Urology, Technical University Munich, Germany
| |
Collapse
|
15
|
Immunological effects of hybrid minimally invasive versus conventional open pancreatoduodenectomy - A single center cohort study. Pancreatology 2021; 21:965-974. [PMID: 33832820 DOI: 10.1016/j.pan.2021.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Minimally invasive surgery is a field of rapid development. Evidence from randomized controlled trials in visceral surgery however still falls short of attesting unequivocal superiority to laparoscopic procedures over conventional open approaches with regard to postoperative outcome. The aim of this study was to explore the perioperative immune status of patients undergoing hybrid minimally invasive or conventional open pancreatoduodenectomy in a prospective cohort study. MATERIAL AND METHODS Subtyping, quantification and functional analysis of circulating immune cells and determination of cytokine-levels in blood samples from patients receiving either hybrid minimally invasive (laPD) or conventional open pancreatoduodenectomy (oPD) was performed. Samples were taken from 29 patients (laPD: n = 14, oPD: n = 15) prior, during and up to six weeks after surgery. Cells were analyzed by flow cytometry, cytokines/chemokines were measured by proximity extension and enzyme-linked immunoassays. RESULTS Open surgery induced higher levels of circulating inflammatory CD14++CD16+ intermediate monocytes. In contrast, hybrid minimally invasive resection was accompanied by increased numbers of circulating regulatory CD4+CD25+CD127low T-cells and by a reduced response of peripheral blood CD3+CD4+ T-cell populations to superantigen stimulation. Yet, rates of postoperative morbidity and infectious complications were similar. CONCLUSIONS In summary, the results of this exploratory study may suggest a more balanced postoperative inflammatory response and a better-preserved immune regulation after hybrid minimally invasive pancreatoduodenectomy when compared to open surgery. Whether these results may translate to or be harnessed for improved patient outcome needs to be determined by future studies including larger cohorts and fully laparoscopic or robotic procedures.
Collapse
|
16
|
Carsetti R, Zaffina S, Piano Mortari E, Terreri S, Corrente F, Capponi C, Palomba P, Mirabella M, Cascioli S, Palange P, Cuccaro I, Milito C, Zumla A, Maeurer M, Camisa V, Vinci MR, Santoro A, Cimini E, Marchioni L, Nicastri E, Palmieri F, Agrati C, Ippolito G, Porzio O, Concato C, Onetti Muda A, Raponi M, Quintarelli C, Quinti I, Locatelli F. Different Innate and Adaptive Immune Responses to SARS-CoV-2 Infection of Asymptomatic, Mild, and Severe Cases. Front Immunol 2020; 11:610300. [PMID: 33391280 PMCID: PMC7772470 DOI: 10.3389/fimmu.2020.610300] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a novel coronavirus, not encountered before by humans. The wide spectrum of clinical expression of SARS-CoV-2 illness suggests that individual immune responses to SARS-CoV-2 play a crucial role in determining the clinical course after first infection. Immunological studies have focused on patients with moderate to severe disease, demonstrating excessive inflammation in tissues and organ damage. In order to understand the basis of the protective immune response in COVID-19, we performed a longitudinal follow-up, flow-cytometric and serological analysis of innate and adaptive immunity in 64 adults with a spectrum of clinical presentations: 28 healthy SARS-CoV-2-negative contacts of COVID-19 cases; 20 asymptomatic SARS-CoV-2-infected cases; eight patients with Mild COVID-19 disease and eight cases of Severe COVID-19 disease. Our data show that high frequency of NK cells and early and transient increase of specific IgA, IgM and, to a lower extent, IgG are associated with asymptomatic SARS-CoV-2 infection. By contrast, monocyte expansion and high and persistent levels of IgA and IgG, produced relatively late in the course of the infection, characterize severe disease. Modest increase of monocytes and different kinetics of antibodies are detected in mild COVID-19. The importance of innate NK cells and the short-lived antibody response of asymptomatic individuals and patients with mild disease suggest that only severe COVID-19 may result in protective memory established by the adaptive immune response.
Collapse
Affiliation(s)
- Rita Carsetti
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eva Piano Mortari
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Sara Terreri
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Francesco Corrente
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Claudia Capponi
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Patrizia Palomba
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Mattia Mirabella
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Simona Cascioli
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paolo Palange
- Department of Public Health and Infectious Diseases Pulmonary Division, Policlinico Umberto I Hospital, Rome, Italy
| | - Ilaria Cuccaro
- Department of Public Health and Infectious Diseases Pulmonary Division, Policlinico Umberto I Hospital, Rome, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alimuddin Zumla
- Center for Clinical Microbiology, Division of Infection and Immunity, University College London, London, United Kingdom
- NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- Immunotherapy Programme, Champalimaud Foundation, Lisbon, Portugal
- Med Clinic, University of Mainz, Mainz, Germany
| | - Vincenzo Camisa
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Maria Rosaria Vinci
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Annapaola Santoro
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology Laboratory, INMI L Spallanzani, IRCCS, Rome, Italy
| | | | | | | | - Chiara Agrati
- Cellular Immunology Laboratory, INMI L Spallanzani, IRCCS, Rome, Italy
| | | | - Ottavia Porzio
- Medical Laboratory Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Concato
- Virology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Andrea Onetti Muda
- Department of Laboratories, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Massimiliano Raponi
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Concetta Quintarelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
| |
Collapse
|
17
|
Kotliar D, Lin AE, Logue J, Hughes TK, Khoury NM, Raju SS, Wadsworth MH, Chen H, Kurtz JR, Dighero-Kemp B, Bjornson ZB, Mukherjee N, Sellers BA, Tran N, Bauer MR, Adams GC, Adams R, Rinn JL, Melé M, Schaffner SF, Nolan GP, Barnes KG, Hensley LE, McIlwain DR, Shalek AK, Sabeti PC, Bennett RS. Single-Cell Profiling of Ebola Virus Disease In Vivo Reveals Viral and Host Dynamics. Cell 2020; 183:1383-1401.e19. [PMID: 33159858 PMCID: PMC7707107 DOI: 10.1016/j.cell.2020.10.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/10/2020] [Accepted: 10/02/2020] [Indexed: 12/14/2022]
Abstract
Ebola virus (EBOV) causes epidemics with high mortality yet remains understudied due to the challenge of experimentation in high-containment and outbreak settings. Here, we used single-cell transcriptomics and CyTOF-based single-cell protein quantification to characterize peripheral immune cells during EBOV infection in rhesus monkeys. We obtained 100,000 transcriptomes and 15,000,000 protein profiles, finding that immature, proliferative monocyte-lineage cells with reduced antigen-presentation capacity replace conventional monocyte subsets, while lymphocytes upregulate apoptosis genes and decline in abundance. By quantifying intracellular viral RNA, we identify molecular determinants of tropism among circulating immune cells and examine temporal dynamics in viral and host gene expression. Within infected cells, EBOV downregulates STAT1 mRNA and interferon signaling, and it upregulates putative pro-viral genes (e.g., DYNLL1 and HSPA5), nominating pathways the virus manipulates for its replication. This study sheds light on EBOV tropism, replication dynamics, and elicited immune response and provides a framework for characterizing host-virus interactions under maximum containment.
Collapse
Affiliation(s)
- Dylan Kotliar
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Aaron E Lin
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Program in Virology, Harvard Medical School, Boston, MA 02115, USA.
| | - James Logue
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Travis K Hughes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemistry, Institute for Medical Engineering and Sciences (IMES), and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Nadine M Khoury
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Siddharth S Raju
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Marc H Wadsworth
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Institute for Medical Engineering and Sciences (IMES), and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Han Chen
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Jonathan R Kurtz
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Bonnie Dighero-Kemp
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Zach B Bjornson
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | | | - Brian A Sellers
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation, National Institutes of Health, Bethesda, MD 20814, USA
| | - Nancy Tran
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Matthew R Bauer
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gordon C Adams
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ricky Adams
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - John L Rinn
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Marta Melé
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia 08034, Spain
| | - Stephen F Schaffner
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Kayla G Barnes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA; MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - Lisa E Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA.
| | - David R McIlwain
- Department of Pathology, Stanford University, Stanford, CA 94305, USA.
| | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemistry, Institute for Medical Engineering and Sciences (IMES), and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Pardis C Sabeti
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Richard S Bennett
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
18
|
Blood Monocyte Subsets with Activation Markers in Relation with Macrophages in Non-Small Cell Lung Cancer. Cancers (Basel) 2020; 12:cancers12092513. [PMID: 32899681 PMCID: PMC7563629 DOI: 10.3390/cancers12092513] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/07/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary This study characterized monocyte subtypes: classical, intermediate, and non-classical with the expression of surfaces markers: CD62L, CD11c, CD18, HLA-DR in non-small cell lung cancer patients (NSCLC) compared to healthy controls and correlations between monocyte subtypes and macrophages in the lung cancer microenvironment. We confirmed the presence of various monocyte subtypes in the blood with predominance of classic monocytes and a higher proportion of classical and intermediate monocytes in NSCLC patients than in healthy subjects. Our observation that intermediate monocytes with CD11c+ and HLA-DR+ expression correlation with the amount of macrophages from the lung cancer microenvironment may indicate role of these cells in cancer immunity. A high proportion of monocytes with low expression of CD62L indicates participation of monocytes in attenuation of anticancer response. The detection and monitoring of the presented monocyte subsets in the blood might be a useful test in lung cancer. Abstract (1) The cells from the monocyte line play an important role as regulators of cancer development and progression. Monocytes present pro- and anti-tumor immunity and differentiation into macrophages. Macrophages are predominant in the lung cancer environment and could be evaluated by bronchoalveolar lavage fluid (BALF). (2) The aim of the study was analysis of monocytes: classical, intermediate and non-classical with expression of: CD62L, CD11c, CD18, HLA-DR in non-small cell lung cancer (NSCLC) and their correlation with BALF macrophages from lungs with cancer (clBALF) and healthy lungs (hlBALF). (3) A total of 24 patients with NSCLC and 20 healthy donors were investigated. Monocyte subtyping and macrophage counts were performed by flow cytometry. (4) There are three types in peripheral blood (PB): classical monocytes (CD14++CD16-), intermediate (CD14+CD16+) and non-classical (CD14-/+CD16++). We noticed a higher proportion of classical and intermediate monocytes in lung cancer than in healthy donors (76.2 vs. 67.3, and 7.9 vs. 5.2 p < 0.05). We observed a higher proportion of macrophages in clBALF then in hlBALF. A higher CD62L expression on all monocyte subtypes in healthy donors than in study group was found. There were positive correlations between: classical CD11c+, intermediate CD11c+, intermediate HLA-DR+ monocytes in PB with macrophages in clBALF. We did not observe these correlations with macrophages from hlBALF. (5) A predominance of classical and intermediate monocytes in lung cancer and the correlation between intermediate monocytes with CD11c+ and HLA-DR+ and macrophages from the NSCLC milieu support a role of monocyte-line cells in cancer immunity. A high proportion of monocytes with low expression of CD62L indicates the participation of monocytes in attenuation of anticancer response.
Collapse
|
19
|
Cormican S, Griffin MD. Human Monocyte Subset Distinctions and Function: Insights From Gene Expression Analysis. Front Immunol 2020; 11:1070. [PMID: 32582174 PMCID: PMC7287163 DOI: 10.3389/fimmu.2020.01070] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/04/2020] [Indexed: 12/30/2022] Open
Abstract
Monocytes are a highly plastic innate immune cell population that displays significant heterogeneity within the circulation. Distinct patterns of surface marker expression have become accepted as a basis for distinguishing three monocyte subsets in humans. These phenotypic subsets, termed classical, intermediate and nonclassical, have also been demonstrated to differ in regard to their functional properties and disease associations when studied in vitro and in vivo. Nonetheless, for the intermediate monocyte subset in particular, functional experiments have yielded conflicting results and some studies point to further levels of heterogeneity. Developments in genetic sequencing technology have provided opportunities to more comprehensively explore the phenotypic and functional differences among conventionally-recognized immune cell subtypes as well as the potential to identify novel subpopulations. In this review, we summarize the transcriptomic evidence in support of the existence of three separate monocyte subsets. We also critically evaluate the insights into subset functional distinctions that have been garnered from monocyte gene expression analysis and the potential utility of such studies to unravel subset-specific functional changes which arise in disease states.
Collapse
Affiliation(s)
- Sarah Cormican
- Regenerative Medical Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.,Nephrology Services, Galway University Hospitals, Saolta University Health Group, Galway, Ireland
| | - Matthew D Griffin
- Regenerative Medical Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.,Nephrology Services, Galway University Hospitals, Saolta University Health Group, Galway, Ireland
| |
Collapse
|
20
|
Girndt M, Trojanowicz B, Ulrich C. Monocytes in Uremia. Toxins (Basel) 2020; 12:toxins12050340. [PMID: 32455723 PMCID: PMC7290468 DOI: 10.3390/toxins12050340] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Monocytes play an important role in both innate immunity and antigen presentation for specific cellular immune defense. In patients with chronic renal failure, as well as those treated with maintenance hemodialysis, these cells are largely dysregulated. There is a large body of literature on monocyte alterations in such patients. However, most of the publications report on small series, there is a vast spectrum of different methods and the heterogeneity of the data prevents any meta-analytic approach. Thus, a narrative review was performed to describe the current knowledge. Monocytes from patients with chronic renal failure differ from those of healthy individuals in the pattern of surface molecule expression, cytokine and mediator production, and function. If these findings can be summarized at all, they might be subsumed as showing chronic inflammation in resting cells together with limited activation upon immunologic challenge. The picture is complicated by the fact that monocytes fall into morphologically and functionally different populations and population shifts interact heavily with dysregulation of the individual cells. Severe complications of chronic renal failure such as impaired immune defense, inflammation, and atherosclerosis can be related to several aspects of monocyte dysfunction. Therefore, this review aims to provide an overview about the impairment and activation of monocytes by uremia and the resulting clinical consequences for renal failure patients.
Collapse
|
21
|
Molecular Mechanisms of Premature Aging in Hemodialysis: The Complex Interplay Between Innate and Adaptive Immune Dysfunction. Int J Mol Sci 2020; 21:ijms21103422. [PMID: 32408613 PMCID: PMC7279398 DOI: 10.3390/ijms21103422] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Hemodialysis (HD) patient are known to be susceptible to a wide range of early and long-term complication such as chronic inflammation, infections, malnutrition, and cardiovascular disease that significantly affect the incidence of mortality. A large gap between the number of people with end-stage kidney disease (ESKD) and patients who received kidney transplantation has been identified. Therefore, there is a huge need to explore the underlying pathophysiology of HD complications in order to provide treatment guidelines. The immunological dysregulation, involving both the innate and adaptive response, plays a crucial role during the HD sessions and in chronic, maintenance treatments. Innate immune system mediators include the dysfunction of neutrophils, monocytes, and natural killer (NK) cells with signaling mediated by NOD-like receptor P3 (NLRP3) and Toll-like receptor 4 (TLR4); in addition, there is a significant activation of the complement system that is mediated by dialysis membrane-surfaces. These effectors induce a persistent, systemic, pro-inflammatory, and pro-coagulant milieu that has been described as inflammaging. The adaptive response, the imbalance in the CD4+/CD8+ T cell ratio, and the reduction of Th2 and regulatory T cells, together with an altered interaction with B lymphocyte by CD40/CD40L, have been mainly implicated in immune system dysfunction. Altogether, these observations suggest that intervention targeting the immune system in HD patients could improve morbidity and mortality. The purpose of this review is to expand our understanding on the role of immune dysfunction in both innate and adaptive response in patients undergoing hemodialysis treatment.
Collapse
|
22
|
Changes in monocyte subsets are associated with clinical outcomes in severe malarial anaemia and cerebral malaria. Sci Rep 2019; 9:17545. [PMID: 31772386 PMCID: PMC6879635 DOI: 10.1038/s41598-019-52579-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/18/2019] [Indexed: 01/17/2023] Open
Abstract
Monocytes are plastic heterogeneous immune cells involved in host-parasite interactions critical for malaria pathogenesis. Human monocytes have been subdivided into three populations based on surface expression of CD14 and CD16. We hypothesised that proportions and phenotypes of circulating monocyte subsets can be markers of severity or fatality in children with malaria. To address this question, we compared monocytes sampled in children with uncomplicated malaria, severe malarial anaemia, or cerebral malaria. Flow cytometry was used to distinguish and phenotype monocyte subsets through CD14, CD16, CD36 and TLR2 expression. Data were first analysed by univariate analysis to evaluate their link to severity and death. Second, multinomial logistic regression was used to measure the specific effect of monocyte proportions and phenotypes on severity and death, after adjustments for other variables unrelated to monocytes. Multivariate analysis demonstrated that decreased percentages of non-classical monocytes were associated with death, suggesting that this monocyte subset has a role in resolving malaria. Using univariate analysis, we also showed that the role of non-classical monocytes involves a mostly anti-inflammatory profile and the expression of CD16. Further studies are needed to decipher the functions of this sub-population during severe malaria episodes, and understand the underlying mechanisms.
Collapse
|
23
|
Changes in the Molecular and Functional Phenotype of Bovine Monocytes during Theileria parva Infection. Infect Immun 2019; 87:IAI.00703-19. [PMID: 31570561 PMCID: PMC6867863 DOI: 10.1128/iai.00703-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 09/21/2019] [Indexed: 12/24/2022] Open
Abstract
Theileria parva is the causative agent of East Coast fever (ECF), a tick-borne disease that kills over a million cattle each year in sub-Saharan Africa. Immune protection against T. parva involves a CD8+ cytotoxic T cell response to parasite-infected cells. However, there is currently a paucity of knowledge regarding the role played by innate immune cells in ECF pathogenesis and T. parva control. Theileria parva is the causative agent of East Coast fever (ECF), a tick-borne disease that kills over a million cattle each year in sub-Saharan Africa. Immune protection against T. parva involves a CD8+ cytotoxic T cell response to parasite-infected cells. However, there is currently a paucity of knowledge regarding the role played by innate immune cells in ECF pathogenesis and T. parva control. Here, we demonstrate an increase in intermediate monocytes (CD14++ CD16+) with a concomitant decrease in the classical (CD14++ CD16−) and nonclassical (CD14+ CD16+) subsets at 12 days postinfection (dpi) during lethal infection but not during nonlethal T. parva infection. Ex vivo analyses of monocytes demonstrated upregulation of interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) mRNA and increased nitric oxide production during T. parva lethal infection compared to nonlethal infection at 10 dpi. Interestingly, no significant differences in peripheral blood parasite loads were observed between lethally and nonlethally infected animals at 12 dpi. In vitro stimulation with T. parva schizont-infected cells or Escherichia coli lipopolysaccharide (LPS) resulted in significant upregulation of IL-1β production by monocytes from lethally infected cattle compared to those from nonlethally infected animals. Strikingly, monocytes from lethally infected animals produced significant amounts of IL-10 mRNA after stimulation with T. parva schizont-infected cells. In conclusion, we demonstrate that T. parva infection leads to alterations in the molecular and functional phenotypes of bovine monocytes. Importantly, since these changes primarily occur in lethal infection, they can serve as biomarkers for ECF progression and severity, thereby aiding in the standardization of protection assessment for T. parva candidate vaccines.
Collapse
|
24
|
New Insights into the Roles of Monocytes/Macrophages in Cardiovascular Calcification Associated with Chronic Kidney Disease. Toxins (Basel) 2019; 11:toxins11090529. [PMID: 31547340 PMCID: PMC6784181 DOI: 10.3390/toxins11090529] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is an important cause of death in patients with chronic kidney disease (CKD), and cardiovascular calcification (CVC) is one of the strongest predictors of CVD in this population. Cardiovascular calcification results from complex cellular interactions involving the endothelium, vascular/valvular cells (i.e., vascular smooth muscle cells, valvular interstitial cells and resident fibroblasts), and monocyte-derived macrophages. Indeed, the production of pro-inflammatory cytokines and oxidative stress by monocyte-derived macrophages is responsible for the osteogenic transformation and mineralization of vascular/valvular cells. However, monocytes/macrophages show the ability to modify their phenotype, and consequently their functions, when facing environmental modifications. This plasticity complicates efforts to understand the pathogenesis of CVC-particularly in a CKD setting, where both uraemic toxins and CKD treatment may affect monocyte/macrophage functions and thereby influence CVC. Here, we review (i) the mechanisms by which each monocyte/macrophage subset either promotes or prevents CVC, and (ii) how both uraemic toxins and CKD therapies might affect these monocyte/macrophage functions.
Collapse
|
25
|
Höpfner F, Jacob M, Ulrich C, Russ M, Simm A, Silber RE, Girndt M, Noutsias M, Werdan K, Schlitt A. Subgroups of monocytes predict cardiovascular events in patients with coronary heart disease. The PHAMOS trial (Prospective Halle Monocytes Study). Hellenic J Cardiol 2019; 60:311-321. [DOI: 10.1016/j.hjc.2019.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/20/2022] Open
|
26
|
Kapellos TS, Bonaguro L, Gemünd I, Reusch N, Saglam A, Hinkley ER, Schultze JL. Human Monocyte Subsets and Phenotypes in Major Chronic Inflammatory Diseases. Front Immunol 2019; 10:2035. [PMID: 31543877 PMCID: PMC6728754 DOI: 10.3389/fimmu.2019.02035] [Citation(s) in RCA: 506] [Impact Index Per Article: 101.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
Human monocytes are divided in three major populations; classical (CD14+CD16−), non-classical (CD14dimCD16+), and intermediate (CD14+CD16+). Each of these subsets is distinguished from each other by the expression of distinct surface markers and by their functions in homeostasis and disease. In this review, we discuss the most up-to-date phenotypic classification of human monocytes that has been greatly aided by the application of novel single-cell transcriptomic and mass cytometry technologies. Furthermore, we shed light on the role of these plastic immune cells in already recognized and emerging human chronic diseases, such as obesity, atherosclerosis, chronic obstructive pulmonary disease, lung fibrosis, lung cancer, and Alzheimer's disease. Our aim is to provide an insight into the contribution of human monocytes to the progression of these diseases and highlight their candidacy as potential therapeutic cell targets.
Collapse
Affiliation(s)
- Theodore S Kapellos
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Lorenzo Bonaguro
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Ioanna Gemünd
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Nico Reusch
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Adem Saglam
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Emily R Hinkley
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| |
Collapse
|
27
|
He T, Xiong J, Huang Y, Zheng C, Liu Y, Bi X, Liu C, Han W, Yang K, Xiao T, Xu X, Yu Y, Huang Y, Zhang J, Zhang B, Zhao J. Klotho restrain RIG-1/NF-κB signaling activation and monocyte inflammatory factor release under uremic condition. Life Sci 2019; 231:116570. [PMID: 31207307 DOI: 10.1016/j.lfs.2019.116570] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/05/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022]
Abstract
AIMS Systemic inflammation is a main hallmark of chronic kidney disease (CKD), but the underlying mechanisms of pathogenesis of CKD-associated systemic inflammation is unclear. Current study was designed to investigate the relationship between indoxyl sulphate (IS) and CKD-associated systemic inflammation along with the protective effects of Klotho in CKD. METHODS IS serum levels from patients were detected by high-performance liquid chromatography (HPLC), and Serum Klotho, IL-6 and TNF-α were measured separately by ELISA and Real-Time PCR analysis. Monocytes were incubated with or without Klotho, while the expressions of retinoic acid-inducible gene I (RIG-I) and NF-κB were analyzed through Western blot assay. Heterozygous kl/kl (kl/+) mice or WT mice were treated with 5/6 renal damage. Thereafter, the CKD mice were intraperitoneally injected with recombinant Klotho protein or PBS. KEY FINDINGS It shows that in 286 CKD patients, the serum levels of inflammatory factors were positively related with IS, but negatively related with Klotho. Klotho significantly inhibited IS-induced RIG-I/NF-κB activation and productions of both IL-6 and TNF-α in cultured monocytes. In vivo, along with the increase of IS and decrease of Klotho in the serum, the activation of RIG-I/NF-κB signaling was observed in peripheral blood monocytes in both CKD mice and patients. Notably, higher levels of IL-6 and TNF-α were detected in kl+/- mice given CKD. Klotho administration has evidently attenuated RIG-I/NF-κB activation in monocytes and systemic inflammation in CKD mice. SIGNIFICANCE The findings suggest that Klotho can suppress CKD-associated systemic inflammation through inhibiting IS-induced RIG-1/NF-κB activation and monocyte inflammatory factor release.
Collapse
Affiliation(s)
- Ting He
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Jiachuan Xiong
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Yinghui Huang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Changling Zheng
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Yong Liu
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Xianjin Bi
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Chi Liu
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Wenhao Han
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Ke Yang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Tangli Xiao
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Xinli Xu
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Yanlin Yu
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Yunjian Huang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Jingbo Zhang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Bo Zhang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China
| | - Jinghong Zhao
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing 400037, PR China.
| |
Collapse
|
28
|
Klingström J, Smed-Sörensen A, Maleki KT, Solà-Riera C, Ahlm C, Björkström NK, Ljunggren HG. Innate and adaptive immune responses against human Puumala virus infection: immunopathogenesis and suggestions for novel treatment strategies for severe hantavirus-associated syndromes. J Intern Med 2019; 285:510-523. [PMID: 30663801 PMCID: PMC6850289 DOI: 10.1111/joim.12876] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Two related hyperinflammatory syndromes are distinguished following infection of humans with hantaviruses: haemorrhagic fever with renal syndrome (HFRS) seen in Eurasia and hantavirus pulmonary syndrome (HPS) seen in the Americas. Fatality rates are high, up to 10% for HFRS and around 35%-40% for HPS. Puumala virus (PUUV) is the most common HFRS-causing hantavirus in Europe. Here, we describe recent insights into the generation of innate and adaptive cell-mediated immune responses following clinical infection with PUUV. First described are studies demonstrating a marked redistribution of peripheral blood mononuclear phagocytes (MNP) to the airways, a process that may underlie local immune activation at the site of primary infection. We then describe observations of an excessive natural killer (NK) cell activation and the persistence of highly elevated numbers of NK cells in peripheral blood following PUUV infection. A similar vigorous CD8 Tcell response is also described, though Tcell responses decline with viraemia. Like MNPs, many NK cells and CD8 T cells also localize to the lung upon acute PUUV infection. Following this, findings demonstrating the ability of hantaviruses, including PUUV, to cause apoptosis resistance in infected target cells, are described. These observations, and associated inflammatory cytokine responses, may provide new insights into HFRS and HPS disease pathogenesis. Based on similarities between inflammatory responses in severe hantavirus infections and other hyperinflammatory disease syndromes, we speculate whether some therapeutic interventions that have been successful in the latter conditions may also be applicable in severe hantavirus infections.
Collapse
Affiliation(s)
- J Klingström
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - K T Maleki
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - C Solà-Riera
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - C Ahlm
- Department of Clinical Microbiology, Infectious Diseases, Umeå University Hospital, Umeå University, Umeå, Sweden
| | - N K Björkström
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - H G Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
29
|
Sabir F, Farooq RK, Asim.ur.Rehman, Ahmed N. Monocyte as an Emerging Tool for Targeted Drug Delivery: A Review. Curr Pharm Des 2019; 24:5296-5312. [DOI: 10.2174/1381612825666190102104642] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/21/2018] [Indexed: 12/30/2022]
Abstract
Monocytes are leading component of the mononuclear phagocytic system that play a key role in phagocytosis and removal of several kinds of microbes from the body. Monocytes are bone marrow precursor cells that stay in the blood for a few days and migrate towards tissues where they differentiate into macrophages. Monocytes can be used as a carrier for delivery of active agents into tissues, where other carriers have no significant access. Targeting monocytes is possible both through passive and active targeting, the former one is simply achieved by enhanced permeation and retention effect while the later one by attachment of ligands on the surface of the lipid-based particulate system. Monocytes have many receptors e.g., mannose, scavenger, integrins, cluster of differentiation 14 (CD14) and cluster of differentiation 36 (CD36). The ligands used against these receptors are peptides, lectins, antibodies, glycolipids, and glycoproteins. This review encloses extensive introduction of monocytes as a suitable carrier system for drug delivery, the design of lipid-based carrier system, possible ways for delivery of therapeutics to monocytes, and the role of monocytes in the treatment of life compromising diseases such as cancer, inflammation, stroke, etc.
Collapse
Affiliation(s)
- Fakhara Sabir
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Rai K. Farooq
- Department of Neuroscience Research, Institute of Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O Box 1982, Dammam 31441, Saudi Arabia
| | - Asim.ur.Rehman
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Naveed Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
30
|
Naicker SD, Cormican S, Griffin TP, Maretto S, Martin WP, Ferguson JP, Cotter D, Connaughton EP, Dennedy MC, Griffin MD. Chronic Kidney Disease Severity Is Associated With Selective Expansion of a Distinctive Intermediate Monocyte Subpopulation. Front Immunol 2018; 9:2845. [PMID: 30619252 PMCID: PMC6302774 DOI: 10.3389/fimmu.2018.02845] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 11/19/2018] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) affects 11–13% of the world's population and greatly increases risk of atherosclerotic cardiovascular disease (ASCVD) and death. It is characterized by systemic inflammation and disturbances in the blood leukocytes that remain incompletely understood. In particular, abnormalities in the numbers and relative proportions of the three major monocyte subsets—classical, intermediate, and non-classical—are described in CKD and end-stage renal disease. In this study, we characterized absolute numbers of blood leukocyte subtypes in adults with renal function varying from normal to advanced CKD. The primary aim was to identify monocyte subpopulations that associated most closely with current estimated glomerular filtration rate (eGFR) and subsequent rate of eGFR decline. Leucocyte and monocyte populations were enumerated by multi-color flow cytometry of whole blood and peripheral blood mononuclear cell (PBMC) samples from adults with CKD stage 1–5 (n = 154) and healthy adults (n = 33). Multiple-linear regression analyses were performed to identify associations between numbers of leucocyte and monocyte populations and clinical characteristics including eGFR and rate of eGFR decline with adjustment for age and gender. In whole blood, total monocyte and neutrophil, but not lymphocyte, numbers were higher in adults with CKD 1-5 compared to no CKD and were significantly associated with current eGFR even following correction for age. In PBMC, classical and intermediate monocyte numbers were higher in CKD 1-5 but only intermediate monocyte numbers were significantly associated with current eGFR in an age-corrected analysis. When intermediate monocytes were further sub-divided into those with mid- and high-level expression of class II MHC (HLA-DRmid and HLA-DRhi intermediate monocytes) it was found that only DRhi intermediate monocytes were increased in number in CKD 1-5 compared to no CKD and were significantly associated with eGFR independently of age among the total (No CKD + CKD 1-5) study cohort as well as those with established CKD (CKD 1-5 only). Furthermore, blood number of DRhi intermediate monocytes alone proved to be significantly associated with subsequent rate of renal functional decline. Together, our data confirm neutrophil and monocyte subset dysregulation in CKD and identify a distinct subpopulation of intermediate monocytes that is associated with higher rate of loss of kidney function.
Collapse
Affiliation(s)
- Serika D Naicker
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - Sarah Cormican
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.,Nephrology Services, Saolta University Healthcare Group, Galway, Ireland
| | - Tomás P Griffin
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.,Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Saolta University Healthcare Group, Galway, Ireland
| | - Silvia Maretto
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - William P Martin
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - John P Ferguson
- HRB Clinical Research Facility, National University of Ireland, Galway, Ireland
| | - Deirdre Cotter
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - Eanna P Connaughton
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - M Conall Dennedy
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - Matthew D Griffin
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.,Nephrology Services, Saolta University Healthcare Group, Galway, Ireland
| |
Collapse
|
31
|
Cold shock Y-box binding protein-1 acetylation status in monocytes is associated with systemic inflammation and vascular damage. Atherosclerosis 2018; 278:156-165. [PMID: 30278358 DOI: 10.1016/j.atherosclerosis.2018.09.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/11/2018] [Accepted: 09/18/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS In dialysis patients, vascular morbidities are highly prevalent and linked to leukocyte extravasation, especially of polarized monocytes. Experimental data demonstrate that phenotypic changes in monocytes require Y-box binding protein-1 (YB-1) upregulation. METHODS We determined YB-1 expression in circulating and vessel-invading monocytes from healthy controls and dialysis patients to correlate results with intima plaque formation and systemic inflammation. RESULTS Compared to healthy subjects, dialysis patients have fewer classical and more intermediate and non-classical monocytes. Post-translationally modified YB-1 (lysine 301/304 acetylation) is detected at high levels in the nucleus of adherent and invading CD14+CD68+ monocytes from umbilical cord and atherosclerosis-prone vessels. The content of non-acetylated YB-1 is significantly decreased (p < 0.001), whereas acetylated YB-1 is correspondingly increased (p < 0.001) throughout all monocyte subpopulations, such that the overall content remains unchanged. CONCLUSIONS In dialysis patients the YB-1 acetylation status is higher with prevailing diabetes and intima plaque formation. Pro-inflammatory mediators TNFα, IL-6, uPAR, CCL2, M-CSF, progranulin, ANP, and midkine, as well as anti-inflammatory IL-10 are significantly increased in dialysis patients, emphasizing a systemic inflammatory milieu. Strong positive correlations of monocytic YB-1 content are seen with ANP, IP-10, IL-6, and IL-10 serum levels. This is the first study demonstrating an association of cold shock protein YB-1 expression with inflammation in hemodialysis patients.
Collapse
|
32
|
Hemodialysis-related changes in phenotypical features of monocytes. Sci Rep 2018; 8:13964. [PMID: 30228352 PMCID: PMC6143543 DOI: 10.1038/s41598-018-31889-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/12/2018] [Indexed: 12/26/2022] Open
Abstract
Hemodialysis (HD) patients exhibit chronic inflammation and leukocyte activation. We investigated the surface-marker profile of monocytes by flow cytometry to assess the chronic effect of uremia and the acute effect of dialysis on their phenotypical and functional features in 16 healthy controls (CON) and 15 HD patients before and after a polysulfone-based dialysis session. Median fluorescence intensities were analyzed indicating expression of CD14, CD16, integrins (CD11b, CD18), chemokine receptors (CCR2, CX3CR1), scavenger receptors (CD36, CD163) and Toll-like receptor-2 (TLR2). Before and after dialysis, HD patients harbour 0.9-fold less CD14++CD16- (Mo1), 1.8-fold more CD14++CD16+ (Mo2) and CD14+CD16++ (Mo3) monocytes than CON. HD patients' Mo1 showed elevated expression of CD11b (1.7-fold), CD18 (1.2-fold) and CD36 (2.1-fold), whereas CD163 expression was reduced in Mo1 and Mo2 (0.6-fold) compared to CON. These markers remained unaffected by dialysis. CX3CR1 expression on Mo2 and Mo3 was lower in HD patients before (0.8-fold) and further diminished after dialysis (0.6-fold). Stimulation of monocytes resulted in diminished responses in HD patients compared to CON. In conclusion, a systematic analysis of the expression of particular surface markers on distinct monocyte subsets may help to distinguish between uremia and/or dialysis induced effects and to evaluate the functionality of monocytes and biocompatibility of HD.
Collapse
|
33
|
Loperena R, Van Beusecum JP, Itani HA, Engel N, Laroumanie F, Xiao L, Elijovich F, Laffer CL, Gnecco JS, Noonan J, Maffia P, Jasiewicz-Honkisz B, Czesnikiewicz-Guzik M, Mikolajczyk T, Sliwa T, Dikalov S, Weyand CM, Guzik TJ, Harrison DG. Hypertension and increased endothelial mechanical stretch promote monocyte differentiation and activation: roles of STAT3, interleukin 6 and hydrogen peroxide. Cardiovasc Res 2018; 114:1547-1563. [PMID: 29800237 PMCID: PMC6106108 DOI: 10.1093/cvr/cvy112] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/11/2018] [Accepted: 05/16/2018] [Indexed: 01/05/2023] Open
Abstract
Aims Monocytes play an important role in hypertension. Circulating monocytes in humans exist as classical, intermediate, and non-classical forms. Monocyte differentiation can be influenced by the endothelium, which in turn is activated in hypertension by mechanical stretch. We sought to examine the role of increased endothelial stretch and hypertension on monocyte phenotype and function. Methods and results Human monocytes were cultured with confluent human aortic endothelial cells undergoing either 5% or 10% cyclical stretch. We also characterized circulating monocytes in normotensive and hypertensive humans. In addition, we quantified accumulation of activated monocytes and monocyte-derived cells in aortas and kidneys of mice with Angiotensin II-induced hypertension. Increased endothelial stretch enhanced monocyte conversion to CD14++CD16+ intermediate monocytes and monocytes bearing the CD209 marker and markedly stimulated monocyte mRNA expression of interleukin (IL)-6, IL-1β, IL-23, chemokine (C-C motif) ligand 4, and tumour necrosis factor α. STAT3 in monocytes was activated by increased endothelial stretch. Inhibition of STAT3, neutralization of IL-6 and scavenging of hydrogen peroxide prevented formation of intermediate monocytes in response to increased endothelial stretch. We also found evidence that nitric oxide (NO) inhibits formation of intermediate monocytes and STAT3 activation. In vivo studies demonstrated that humans with hypertension have increased intermediate and non-classical monocytes and that intermediate monocytes demonstrate evidence of STAT3 activation. Mice with experimental hypertension exhibit increased aortic and renal infiltration of monocytes, dendritic cells, and macrophages with activated STAT3. Conclusions These findings provide insight into how monocytes are activated by the vascular endothelium during hypertension. This is likely in part due to a loss of NO signalling and increased release of IL-6 and hydrogen peroxide by the dysfunctional endothelium and a parallel increase in STAT activation in adjacent monocytes. Interventions to enhance bioavailable NO, reduce IL-6 or hydrogen peroxide production or to inhibit STAT3 may have anti-inflammatory roles in hypertension and related conditions.
Collapse
Affiliation(s)
- Roxana Loperena
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Justin P Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hana A Itani
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Noah Engel
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Fanny Laroumanie
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liang Xiao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juan S Gnecco
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Jonathan Noonan
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Barbara Jasiewicz-Honkisz
- Department of Internal Medicine, Jagiellonian University School of Medicine, Cracow, Poland
- Department of Immunology, Jagiellonian University School of Medicine, Cracow, Poland
| | | | - Tomasz Mikolajczyk
- Department of Internal Medicine, Jagiellonian University School of Medicine, Cracow, Poland
- Department of Immunology, Jagiellonian University School of Medicine, Cracow, Poland
| | - Tomasz Sliwa
- Department of Internal Medicine, Jagiellonian University School of Medicine, Cracow, Poland
- Department of Immunology, Jagiellonian University School of Medicine, Cracow, Poland
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - David G Harrison
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
34
|
Alahakoon TI, Medbury H, Williams H, Fewings N, Wang XM, Lee VW. Distribution of monocyte subsets and polarization in preeclampsia and intrauterine fetal growth restriction. J Obstet Gynaecol Res 2018; 44:2135-2148. [PMID: 30058176 DOI: 10.1111/jog.13770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/04/2018] [Indexed: 12/20/2022]
Abstract
AIM Monocytes are likely to play a significant role in the pathogenesis of preeclampsia (PE) and intrauterine fetal growth restriction (IUGR), given their role in homeostasis and tissue repair. Our aim was to study the gestational changes in monocytes in normal pregnancy and to determine whether monocyte subsets and phenotype are altered in pregnancy complications, such as PE and IUGR. METHODS A prospective cross-sectional case-control study was conducted. Pregnant women between 24 and 40 weeks of gestation (n = 54) were recruited and classified into four clinical groups of normal pregnancy, PE, IUGR and PE + IUGR. The maternal monocyte subsets classical, intermediate and nonclassical were compared for each clinical group. Monocyte polarization towards M1 (inflammatory) and M2 (repair) phenotypes was assessed by surface expression of CD86 and CD163 ratio, using flow cytometry. RESULTS The classical monocytes were reduced and intermediate monocyte elevated compared to normal pregnancy in PE, IUGR and PE + IUGR in gestations <37 weeks and IUGR in 26-40 weeks. CD163 expression was increased and CD86/CD163 ratio decreased in IUGR compared to normal pregnancy for all subsets. Nonclassical monocyte counts and CD163 expression increased with advancing gestation in normal pregnancy. CONCLUSION These results show for the first time, a shift towards increased intermediate maternal monocyte subtype in IUGR and in preterm PE as well as skewing of maternal peripheral monocytes (all subsets) towards M2 phenotype in pregnancies complicated by IUGR.
Collapse
Affiliation(s)
- Thushari I Alahakoon
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Westmead Institute for Maternal and Fetal Medicine, Westmead Hospital, Westmead, New South Wales, Australia
| | - Heather Medbury
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Surgery, Westmead Hospital, Westmead, New South Wales, Australia
| | - Helen Williams
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Surgery, Westmead Hospital, Westmead, New South Wales, Australia
| | - Nicole Fewings
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Immunology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Xin M Wang
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Flow Cytometry Core Facility, Westmead Research Hub, Westmead, New South Wales, Australia
| | - Vincent W Lee
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia
| |
Collapse
|
35
|
Cornwell WD, Kim V, Fan X, Vega ME, Ramsey FV, Criner GJ, Rogers TJ. Activation and polarization of circulating monocytes in severe chronic obstructive pulmonary disease. BMC Pulm Med 2018; 18:101. [PMID: 29907106 PMCID: PMC6003040 DOI: 10.1186/s12890-018-0664-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/29/2018] [Indexed: 01/03/2023] Open
Abstract
Background The ability of circulating monocytes to develop into lung macrophages and promote lung tissue damage depends upon their phenotypic pattern of differentiation and activation. Whether this phenotypic pattern varies with COPD severity is unknown. Here we characterize the activation and differentiation status of circulating monocytes in patients with moderate vs. severe COPD. Methods Blood monocytes were isolated from normal non-smokers (14), current smokers (13), patients with moderate (9), and severe COPD (11). These cells were subjected to analysis by flow cytometry to characterize the expression of activation markers, chemoattractant receptors, and surface markers characteristic of either M1- or M2-type macrophages. Results Patients with severe COPD had increased numbers of total circulating monocytes and non-classical patrolling monocytes, compared to normal subjects and patients with moderate COPD. In addition, while the percentage of circulating monocytes that expressed an M2-like phenotype was reduced in patients with either moderate or severe disease, the levels of expression of M2 markers on this subpopulation of monocytes in severe COPD was significantly elevated. This was particularly evident for the expression of the chemoattractant receptor CCR5. Conclusions Blood monocytes in severe COPD patients undergo unexpected pre-differentiation that is largely characteristic of M2-macrophage polarization, leading to the emergence of an unusual M2-like monocyte population with very high levels of CCR5. These results show that circulating monocytes in patients with severe COPD possess a cellular phenotype which may permit greater mobilization to the lung, with a pre-existing bias toward a potentially destructive inflammatory phenotype. Electronic supplementary material The online version of this article (10.1186/s12890-018-0664-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William D Cornwell
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA. .,Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| | - Victor Kim
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Xiaoxuan Fan
- Temple University Flow Cytometry Facility, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Marie Elena Vega
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Frederick V Ramsey
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Gerard J Criner
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.,Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.,Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
36
|
de Martini CC, de Andrade JT, de Almeida SKM, Oliveira Silva KL, de Rezende Eugenio F, Dos Santos PSP, de Lima VMF. Cellular apoptosis and nitric oxide production in PBMC and spleen from dogs with visceral leishmaniasis. Comp Immunol Microbiol Infect Dis 2018; 57:1-7. [PMID: 30017072 DOI: 10.1016/j.cimid.2018.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/25/2022]
Abstract
Nitric oxide (NO) is involved in the death of the Leishmania parasite and regulation of apoptosis. We quantified the frequency of cells producing NO and its levels in the peripheral blood mononuclear cells (PBMC), leukocytes from spleen in Visceral Leishmaniasis (VL) symptomatic dogs and correlated NO levels with apoptosis and parasite load in the spleen. The percentage of NO+ cells and CD14+/NO+ was higher in PBMC and spleen cells in infected dogs than in controls. The levels of NO+ and CD14+/NO+ cells was higher in PBMC, but lower spleen of dogs infected than compared to control. Late apoptosis rates increased in PBMC and spleen of infected dogs compared to controls, and the NO levels and apoptosis not showed correlation. There was a positive correlation between the percentage of cells producing NO in the spleen and parasite load. The NO participates in the immune response in the canine VL, but it is not apoptosis inducer.
Collapse
Affiliation(s)
- Cleber Costa de Martini
- Department of Clinical Care, Surgery and Animal Reproduction, Laboratory of Cellular Immunology, Faculty of Veterinary Medicine of Araçatuba, Universidade Estadual Paulista, "Julio de Mesquita Filho", FMVA/UNESP, Rua Clovis Pestana, 793, Araçatuba, São Paulo, CEP 16050-680, Brazil
| | - Jéssica Thomé de Andrade
- Department of Clinical Care, Surgery and Animal Reproduction, Laboratory of Cellular Immunology, Faculty of Veterinary Medicine of Araçatuba, Universidade Estadual Paulista, "Julio de Mesquita Filho", FMVA/UNESP, Rua Clovis Pestana, 793, Araçatuba, São Paulo, CEP 16050-680, Brazil
| | - Stéfani Karin Martiniano de Almeida
- Department of Clinical Care, Surgery and Animal Reproduction, Laboratory of Cellular Immunology, Faculty of Veterinary Medicine of Araçatuba, Universidade Estadual Paulista, "Julio de Mesquita Filho", FMVA/UNESP, Rua Clovis Pestana, 793, Araçatuba, São Paulo, CEP 16050-680, Brazil
| | - Kathlenn Liezbeth Oliveira Silva
- Department of Clinical Care, Surgery and Animal Reproduction, Laboratory of Cellular Immunology, Faculty of Veterinary Medicine of Araçatuba, Universidade Estadual Paulista, "Julio de Mesquita Filho", FMVA/UNESP, Rua Clovis Pestana, 793, Araçatuba, São Paulo, CEP 16050-680, Brazil
| | - Flavia de Rezende Eugenio
- Department of Clinical Care, Surgery and Animal Reproduction, Laboratory of Cellular Immunology, Faculty of Veterinary Medicine of Araçatuba, Universidade Estadual Paulista, "Julio de Mesquita Filho", FMVA/UNESP, Rua Clovis Pestana, 793, Araçatuba, São Paulo, CEP 16050-680, Brazil
| | - Paulo Sergio Patto Dos Santos
- Department of Clinical Care, Surgery and Animal Reproduction, Laboratory of Cellular Immunology, Faculty of Veterinary Medicine of Araçatuba, Universidade Estadual Paulista, "Julio de Mesquita Filho", FMVA/UNESP, Rua Clovis Pestana, 793, Araçatuba, São Paulo, CEP 16050-680, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Clinical Care, Surgery and Animal Reproduction, Laboratory of Cellular Immunology, Faculty of Veterinary Medicine of Araçatuba, Universidade Estadual Paulista, "Julio de Mesquita Filho", FMVA/UNESP, Rua Clovis Pestana, 793, Araçatuba, São Paulo, CEP 16050-680, Brazil.
| |
Collapse
|
37
|
Pino CJ, Westover AJ, Johnston KA, Buffington DA, Humes HD. Regenerative Medicine and Immunomodulatory Therapy: Insights From the Kidney, Heart, Brain, and Lung. Kidney Int Rep 2018; 3:771-783. [PMID: 29989023 PMCID: PMC6035130 DOI: 10.1016/j.ekir.2017.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/21/2017] [Accepted: 12/31/2017] [Indexed: 12/30/2022] Open
Abstract
Regenerative medicine was initially focused on tissue engineering to replace damaged tissues and organs with constructs derived from cells and biomaterials. More recently, this field of inquiry has expanded into exciting areas of translational medicine modulating the body’s own endogenous processes, to prevent tissue damage in organs and to repair and regenerate these damaged tissues. This review will focus on recent insights derived from studies in which the manipulation of the innate immunologic system may diminish acute kidney injury and enhance renal repair and recovery without the progression to chronic kidney disease and renal failure. The manner in which these interventions may improve acute and chronic organ dysfunction, including the heart, brain, and lung, will also be reviewed.
Collapse
Affiliation(s)
| | | | | | | | - H David Humes
- Innovative BioTherapies, Inc., Ann Arbor, Michigan, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.,CytoPherx, Inc., Ann Arbor, Michigan, USA
| |
Collapse
|
38
|
Immunomodulatory Device Promotes a Shift of Circulating Monocytes to a Less Inflammatory Phenotype in Chronic Hemodialysis Patients. ASAIO J 2017; 62:623-30. [PMID: 27258222 DOI: 10.1097/mat.0000000000000400] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Patients with end-stage renal disease (ESRD) on chronic hemodialysis (HD) suffer accelerated morbidity and mortality rates caused by cardiovascular disease and infections. Chronic inflammation plays a critical role in these poor outcomes. The activated monocyte (MO) has become a prime therapeutic target to modulate this inflammatory process. A selective cytopheretic device (SCD) was evaluated to assess its effects on the circulating MO pool. A pilot trial was undertaken in 15 ESRD patients on HD with C-reactive protein (CRP) levels greater than 5 mg/dl. An excellent safety profile was observed with no decline in leukocyte (LE) or platelet counts. The effect of SCD therapy on MO phenotypes in these patients was determined on peripheral blood MO utilizing flow cytometry. SCD therapy promoted a shift in MO phenotype from predominantly CD14 expressing MO at baseline/pre-SCD therapy to CD14 expressing MO post-SCD therapy. A significant shift in MO population phenotype afforded by a single SCD therapy session was observed (p < 0.013). In a subset of patients (n = 7) presenting with type 2 diabetes mellitus (T2D), this persistent decline in MO CD14 expression was sustained as long as 2 weeks posttherapy. These results demonstrate that the SCD therapy has the potential to modulate the chronic proinflammatory state in ESRD patients.
Collapse
|
39
|
Bomberg H, Volk T, Biedler A, Schneider SO. Impact of intraoperative blood salvage on monocyte subsets alteration and intracellular tumor necrosis factor-α production. J Biomed Mater Res A 2017; 106:815-821. [PMID: 29094483 DOI: 10.1002/jbm.a.36281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 11/09/2022]
Abstract
Intraoperative salvaged blood is used to reduce allogeneic blood transfusion in orthopedic surgery patients. However, salvaged blood reinfusion may lead to immune reactions. Salvaged and venous blood from 20 patients undergoing hip arthroplasty was processed. The salvaged samples were mixed with patients' venous blood and incubated in absence or presence of lipopolysaccharide. SAMPLES Venous: venous patient blood (n = 20). Native: mixed salvaged native blood (n = 20). Filtered: mixed salvaged leukocyte filtered blood (n = 20). Irradiated: mixed salvaged irradiated blood (n = 20). The frequency of the surface receptors CD14, HLA-DR, and intracellular tumor necrosis factor (TNF)-α on peripheral blood mononuclear cells was analyzed by fluorescence-activated cell sorting analysis. The frequency of unstimulated CD14low and CD14high cells as well as unstimulated HLA-DR and TNF-α positive monocytes was comparable between venous and filtered salvaged blood. However, native and irradiated salvaged blood increased compared with venous (p < 0.05) and filtered salvaged blood (p < 0.05) for unstimulated CD14low cells, HLA-DR, and TNF-α positive monocytes. Stimulated intracellular TNF-α positive monocytes were decreased in native, filtered, and irradiated salvaged blood compared with venous blood (p < 0.05). Processing perioperative salvaged blood with leukofiltration minimizes the influence on monocytes activation compared with native and irradiated salvaged blood. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 815-821, 2018.
Collapse
Affiliation(s)
- Hagen Bomberg
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Saarland University Medical Center, Homburg, Saar, Germany
| | - Thomas Volk
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Saarland University Medical Center, Homburg, Saar, Germany
| | - Andreas Biedler
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Saarland University Medical Center, Homburg, Saar, Germany
| | - Sven O Schneider
- Department of Anesthesiology, Intensive Care Medicine and Pain Medicine, Saarland University Medical Center, Homburg, Saar, Germany
| |
Collapse
|
40
|
Proportions of Proinflammatory Monocytes Are Important Predictors of Mortality Risk in Hemodialysis Patients. Mediators Inflamm 2017; 2017:1070959. [PMID: 29200664 PMCID: PMC5671738 DOI: 10.1155/2017/1070959] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 08/06/2017] [Accepted: 09/13/2017] [Indexed: 11/25/2022] Open
Abstract
Despite the continuous progression in dialysis medicine, mortality and the burden of cardiovascular disease (CVD) among hemodialysis patients are still substantial. Substantial evidence suggests that proinflammatory (CD16+) monocytes contribute to the development of atherosclerosis. A cohort of 136 stable hemodialysis patients (follow-up: 6.25 year) was assessed to investigate the association between the proportion of CD16+ monocytes for all-cause and CVD mortalities. The CD16+ monocytes were associated with both mortalities after adjusting for a preexisting CVD history. Compared to the reference group (CD16+ monocytes within [15.6–18.6], the first and second quartile), patients with CD16+ monocytes above the highest quartile level (>21.5) had an adjusted hazard ratio (HR) of 30.85 (95% confidence interval [CI]: 7.12–133.8) for CVD mortality and 5.28 (2.07–13.49) for all-cause mortality, and those with CD16+ monocytes below the lowest quartile ≤15.6), had significantly elevated death risks after 3.5-year follow-up (HR [95% CI]: 10.9 [2.42–48.96] and 4.38 [1.45–13.24] for CV and all-cause mortalities, respectively). The hemodialysis patients with CD16+ monocyte level in a low but mostly covering normal range also portended a poor prognosis. The findings shed some light for nephrologists on future prospects of early recognizing immune dysfunction and improving early intervention outcomes.
Collapse
|
41
|
Tang Y, Joyner CJ, Cabrera-Mora M, Saney CL, Lapp SA, Nural MV, Pakala SB, DeBarry JD, Soderberg S, Kissinger JC, Lamb TJ, Galinski MR, Styczynski MP. Integrative analysis associates monocytes with insufficient erythropoiesis during acute Plasmodium cynomolgi malaria in rhesus macaques. Malar J 2017; 16:384. [PMID: 28938907 PMCID: PMC5610412 DOI: 10.1186/s12936-017-2029-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/12/2017] [Indexed: 01/06/2023] Open
Abstract
Background Mild to severe anaemia is a common complication of malaria that is caused in part by insufficient erythropoiesis in the bone marrow. This study used systems biology to evaluate the transcriptional and alterations in cell populations in the bone marrow during Plasmodium cynomolgi infection of rhesus macaques (a model of Plasmodium vivax malaria) that may affect erythropoiesis. Results An appropriate erythropoietic response did not occur to compensate for anaemia during acute cynomolgi malaria despite an increase in erythropoietin levels. During this period, there were significant perturbations in the bone marrow transcriptome. In contrast, relapses did not induce anaemia and minimal changes in the bone marrow transcriptome were detected. The differentially expressed genes during acute infection were primarily related to ongoing inflammatory responses with significant contributions from Type I and Type II Interferon transcriptional signatures. These were associated with increased frequency of intermediate and non-classical monocytes. Recruitment and/or expansion of these populations was correlated with a decrease in the erythroid progenitor population during acute infection, suggesting that monocyte-associated inflammation may have contributed to anaemia. The decrease in erythroid progenitors was associated with downregulation of genes regulated by GATA1 and GATA2, two master regulators of erythropoiesis, providing a potential molecular basis for these findings. Conclusions These data suggest the possibility that malarial anaemia may be driven by monocyte-associated disruption of GATA1/GATA2 function in erythroid progenitors resulting in insufficient erythropoiesis during acute infection. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-2029-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Tang
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Chester J Joyner
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Monica Cabrera-Mora
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Celia L Saney
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Stacey A Lapp
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Mustafa V Nural
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA.,Department of Computer Science, University of Georgia, Athens, GA, USA
| | - Suman B Pakala
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Jeremy D DeBarry
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Stephanie Soderberg
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | - Jessica C Kissinger
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA.,Department of Genetics, University of Georgia, Athens, GA, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA.,Department of Computer Science, University of Georgia, Athens, GA, USA
| | - Tracey J Lamb
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Mary R Galinski
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Mark P Styczynski
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA. .,Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
| |
Collapse
|
42
|
Sugimoto C, Merino KM, Hasegawa A, Wang X, Alvarez XA, Wakao H, Mori K, Kim WK, Veazey RS, Didier ES, Kuroda MJ. Critical Role for Monocytes/Macrophages in Rapid Progression to AIDS in Pediatric Simian Immunodeficiency Virus-Infected Rhesus Macaques. J Virol 2017; 91:e00379-17. [PMID: 28566378 PMCID: PMC5553179 DOI: 10.1128/jvi.00379-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/22/2017] [Indexed: 12/23/2022] Open
Abstract
Infant humans and rhesus macaques infected with the human or simian immunodeficiency virus (HIV or SIV), respectively, express higher viral loads and progress more rapidly to AIDS than infected adults. Activated memory CD4+ T cells in intestinal tissues are major primary target cells for SIV/HIV infection, and massive depletion of these cells is considered a major cause of immunodeficiency. Monocytes and macrophages are important cells of innate immunity and also are targets of HIV/SIV infection. We reported previously that a high peripheral blood monocyte turnover rate was predictive for the onset of disease progression to AIDS in SIV-infected adult macaques. The purpose of this study was to determine if earlier or higher infection of monocytes/macrophages contributes to the more rapid progression to AIDS in infants. We observed that uninfected infant rhesus macaques exhibited higher physiologic baseline monocyte turnover than adults. Early after SIV infection, the monocyte turnover further increased, and it remained high during progression to AIDS. A high percentage of terminal deoxynucleotidyltransferase dUTP nick end label (TUNEL)-positive macrophages in the lymph nodes (LNs) and intestine corresponded with an increasing number of macrophages derived from circulating monocytes (bromodeoxyuridine positive [BrdU+] CD163+), suggesting that the increased blood monocyte turnover was required to rapidly replenish destroyed tissue macrophages. Immunofluorescence analysis further demonstrated that macrophages were a significant portion of the virus-producing cells found in LNs, intestinal tissues, and lungs. The higher baseline monocyte turnover in infant macaques and subsequent macrophage damage by SIV infection may help explain the basis of more rapid disease progression to AIDS in infants.IMPORTANCE HIV infection progresses much more rapidly in pediatric cases than in adults; however, the mechanism for this difference is unclear. Using the rhesus macaque model, this work was performed to address why infants infected with SIV progress more quickly to AIDS than do adults. Earlier we reported that in adult rhesus macaques, increasing monocyte turnover reflected tissue macrophage damage by SIV and was predictive of terminal disease progression to AIDS. Here we report that uninfected infant rhesus macaques exhibited a higher physiological baseline monocyte turnover rate than adults. Furthermore, once infected with SIV, infants displayed further increased monocyte turnover that may have facilitated the accelerated progression to AIDS. These results support a role for monocytes and macrophages in the pathogenesis of SIV/HIV and begin to explain why infants are more prone to rapid disease progression.
Collapse
Affiliation(s)
- Chie Sugimoto
- Division of Immunology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Kristen M Merino
- Division of Immunology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Atsuhiko Hasegawa
- Division of Immunology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Xiaolei Wang
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Xavier A Alvarez
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Hiroshi Wakao
- Department of Hygiene and Cellular Preventive Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Kazuyasu Mori
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Ronald S Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Elizabeth S Didier
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, USA
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Marcelo J Kuroda
- Division of Immunology, Tulane National Primate Research Center, Covington, Louisiana, USA
| |
Collapse
|
43
|
Scholz S, Baharom F, Rankin G, Maleki KT, Gupta S, Vangeti S, Pourazar J, Discacciati A, Höijer J, Bottai M, Björkström NK, Rasmuson J, Evander M, Blomberg A, Ljunggren HG, Klingström J, Ahlm C, Smed-Sörensen A. Human hantavirus infection elicits pronounced redistribution of mononuclear phagocytes in peripheral blood and airways. PLoS Pathog 2017. [PMID: 28640917 PMCID: PMC5498053 DOI: 10.1371/journal.ppat.1006462] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hantaviruses infect humans via inhalation of virus-contaminated rodent excreta. Infection can cause severe disease with up to 40% mortality depending on the viral strain. The virus primarily targets the vascular endothelium without direct cytopathic effects. Instead, exaggerated immune responses may inadvertently contribute to disease development. Mononuclear phagocytes (MNPs), including monocytes and dendritic cells (DCs), orchestrate the adaptive immune responses. Since hantaviruses are transmitted via inhalation, studying immunological events in the airways is of importance to understand the processes leading to immunopathogenesis. Here, we studied 17 patients infected with Puumala virus that causes a mild form of hemorrhagic fever with renal syndrome (HFRS). Bronchial biopsies as well as longitudinal blood draws were obtained from the patients. During the acute stage of disease, a significant influx of MNPs expressing HLA-DR, CD11c or CD123 was detected in the patients’ bronchial tissue. In parallel, absolute numbers of MNPs were dramatically reduced in peripheral blood, coinciding with viremia. Expression of CCR7 on the remaining MNPs in blood suggested migration to peripheral and/or lymphoid tissues. Numbers of MNPs in blood subsequently normalized during the convalescent phase of the disease when viral RNA was no longer detectable in plasma. Finally, we exposed blood MNPs in vitro to Puumala virus, and demonstrated an induction of CCR7 expression on MNPs. In conclusion, the present study shows a marked redistribution of blood MNPs to the airways during acute hantavirus disease, a process that may underlie the local immune activation and contribute to immunopathogenesis in hantavirus-infected patients. Inhalation of hantavirus-infected rodent droppings can cause a wide range of disease ranging from mild symptoms to deaths in humans. Central to hantavirus disease is vascular leakage that can manifest in different organs, including the lungs. Although the virus can infect endothelial cells lining the blood vessels, it does not cause cell death. Instead, activation of the immune system in response to viral infection has been implicated in causing vascular leakage. In this study, we investigated how monocytes and dendritic cells (DCs) are involved in hantavirus disease, given their capacity to activate other immune cells. We obtained unique clinical material from 17 Puumala virus-infected patients including mucosal biopsies from the airways as well as multiple blood draws over the course of disease. In the airways of these patients, we observed an infiltration of monocytes and DCs. In parallel, there was a dramatic depletion in peripheral blood—more than ten-fold—of monocytes and DCs that was sustained throughout the first two weeks of disease. Taken together, this study provides novel insights into immune mediated processes underlying human hantavirus pathogenesis.
Collapse
Affiliation(s)
- Saskia Scholz
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Faezzah Baharom
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Gregory Rankin
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Kimia T. Maleki
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Shawon Gupta
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sindhu Vangeti
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jamshid Pourazar
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Andrea Discacciati
- Unit of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Höijer
- Unit of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matteo Bottai
- Unit of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Niklas K. Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Rasmuson
- Department of Clinical Microbiology, Infectious Diseases, Umeå University, Umeå, Sweden
| | - Magnus Evander
- Department of Clinical Microbiology, Virology, Umeå University, Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Clas Ahlm
- Department of Clinical Microbiology, Infectious Diseases, Umeå University, Umeå, Sweden
| | - Anna Smed-Sörensen
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
44
|
Kolseth IBM, Reine TM, Parker K, Sudworth A, Witczak BJ, Jenssen TG, Kolset SO. Increased levels of inflammatory mediators and proinflammatory monocytes in patients with type I diabetes mellitus and nephropathy. J Diabetes Complications 2017; 31:245-252. [PMID: 27452162 DOI: 10.1016/j.jdiacomp.2016.06.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/20/2016] [Accepted: 06/28/2016] [Indexed: 12/31/2022]
Abstract
AIMS To investigate and describe the relationship between diabetic nephropathy and systemic inflammation in patients with type 1 diabetes mellitus (T1DM). METHODS Patients with T1DM, with or without reduced renal function due to diabetic nephropathy, were included. Differences in inflammatory mediators, adhesion molecules, markers of endothelial dysfunction and subsets of monocytes were studied in patients with mean disease duration of 31years. RESULTS Patients with T1DM with and without renal failure were compared. Patients with nephropathy had increased plasma levels of proinflammatory monocytes, as well as circulatory PAI-1, syndecan-1, VEGF, IL-1β, IL-1Ra and CCL4. Peripheral blood mononuclear cells from patients with nephropathy numerically increased soluble ICAM and PAI-1 in co-culture with primary endothelial cells compared to cells from patients without nephropathy. CONCLUSIONS T1DM patients with kidney failure have higher levels of proinflammatory monocytes and circulatory inflammatory mediators compared to patients with T1DM alone. The results highlight the importance of inflammation and endothelial dysfunction in diabetic nephropathy with reduced GFR.
Collapse
MESH Headings
- Biomarkers/blood
- Cells, Cultured
- Diabetes Mellitus, Type 1/complications
- Diabetic Angiopathies/blood
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diabetic Nephropathies/blood
- Diabetic Nephropathies/immunology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Disease Progression
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Human Umbilical Vein Endothelial Cells/cytology
- Human Umbilical Vein Endothelial Cells/immunology
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Inflammation Mediators/blood
- Inflammation Mediators/metabolism
- Intercellular Adhesion Molecule-1/blood
- Intercellular Adhesion Molecule-1/metabolism
- Kidney Failure, Chronic/complications
- Kidney Failure, Chronic/immunology
- Kidney Failure, Chronic/metabolism
- Kidney Failure, Chronic/pathology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/pathology
- Male
- Middle Aged
- Monocytes/immunology
- Monocytes/metabolism
- Monocytes/pathology
- Plasminogen Activator Inhibitor 1/blood
- Plasminogen Activator Inhibitor 1/metabolism
- Renal Insufficiency/complications
- Renal Insufficiency/immunology
- Renal Insufficiency/metabolism
- Renal Insufficiency/pathology
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/immunology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Severity of Illness Index
- Up-Regulation
Collapse
Affiliation(s)
| | - Trine Marita Reine
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Krystina Parker
- Department of Nephrology, Akershus University Hospital, Norway
| | - Amanda Sudworth
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Bartlomiej J Witczak
- Department of Nephrology, Akershus University Hospital, Norway; Department of Nephrology, Oslo University Hospital, Ullevaal, Oslo, Norway
| | - Trond Geir Jenssen
- Department of Transplant Medicine, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; The Metabolic and Renal Research Group, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Svein Olav Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
45
|
Bargalló ME, Guardo AC, Maleno MJ, Miralles L, Egaña-Gorroño L, Escribà T, García F, Gatell JM, Arnedo M, Plana M. Utility of Systematic Isolation of immune cell subsets from HIV-infected individuals for miRNA profiling. J Immunol Methods 2016; 442:12-19. [PMID: 28039100 DOI: 10.1016/j.jim.2016.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/15/2016] [Accepted: 12/15/2016] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Peripheral blood mononuclear cells (PBMCs) are frequently used for genomic analyses, but several factors can affect the yield and integrity of nucleic acids, including the methods of cell collection and isolation. The goal of this work was to analyze the utility of systematic isolation of different immune cell subsets by immunomagnetic separation and the RNA integrity after isolated cells from samples of HIV-infected patients. METHODS PBMC from Healthy Controls (HC, n=15), Elite Controllers (EC, n=15), Viremic Controllers (VC, n=15), Viremic Progressors (VP, n=15) and HIV-infected patients on therapy (ART, n=15) were isolated by Ficoll-Paque density gradient centrifugation. Subsets were separated with monoclonal antibodies (CD56, CD14, CD4, and CD8) conjugated to microbeads. We evaluated the yield and purity of each subset isolated from PBMCs under resting and activated conditions; LPS, anti-CD3/CD28 and anti-CD16 were used to activate monocytes, PBMC, T cells and NK cells, respectively. The quality of extracted RNA was tested by 2100 Bioanalyzer. RESULTS In resting conditions, the average yield of CD14+ (monocytes) was decreased (p=0.021) in HIV+ patients compared with healthy controls. CD56+ (Natural Killer-NKs; p=0.03) and CD8+ (Cytotoxic T lymphocytes-CTL p=0.001) cells were increased in HIV+ patients after 72h of activation. The purity assay detected significant differences in CD14+ (p≤0.001) and CD8+ (p=0.034) subpopulations when comparing PBMC isolated either from healthy controls or HIV+ patients. The number of activated cells in HIV+ presented differences in CD8 subset (p=0.003). Finally, similar quantities of high quality RNA were extracted from immune cells subsets obtained by our method. Specifically, we show that Bioanalyzer electrophenograms reveal optimal RIN values in HIV positive and negative patients in resting condition (EC:8;HC:6.5;VC:8.80;VP:8;HAART:7.5) and activated condition (EC:9;HC:6.7;VC:8.2;VP:7.2;HAART:8.6). CONCLUSION This method allowed us to obtain a sufficient quantity of different isolated immune cell subsets from HIV-infected individuals at different disease stages. Moreover, the assessed qualities of nucleic acids allow us to perform subsequent molecular studies, such as microRNA profiling.
Collapse
Affiliation(s)
- Manel E Bargalló
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Alberto C Guardo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Maria J Maleno
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Laia Miralles
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lander Egaña-Gorroño
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Tuixent Escribà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Felipe García
- Infectious Diseases Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Jose M Gatell
- Infectious Diseases Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Mireia Arnedo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Montserrat Plana
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic de Barcelona, Barcelona, Spain.
| |
Collapse
|
46
|
Xing Z, Han J, Hao X, Wang J, Jiang C, Hao Y, Wang H, Wu X, Shen L, Dong X, Li T, Li G, Zhang J, Hou X, Zeng H. Immature monocytes contribute to cardiopulmonary bypass-induced acute lung injury by generating inflammatory descendants. Thorax 2016; 72:245-255. [PMID: 27660037 DOI: 10.1136/thoraxjnl-2015-208023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND As immune regulatory and effector cells, monocytes play an important role in the blood-extracorporeal circuit contact-related acute lung injury in patients undergoing cardiopulmonary bypass (CPB). However, circulating monocytes are phenotypically and functionally heterogeneous, so we characterised how immature monocytes affect acute lung injury induced by CPB. METHODS The identification and dynamic changes in monocyte subsets were monitored by flow cytometry in patients undergoing CPB and in a rat model of CPB. The differentiation and migration of monocyte subsets were explored by in vitro cultures and adoptive transfer in the CPB rat model. RESULTS We observed a dramatic increase of two monocyte subsets in the peripheral blood of patients undergoing CPB, involving tumour necrosis factor (TNF)-α-producing, mature intermediate CD14highCD16+ monocytes and a novel immature CD14lowCD16- subset. The immature CD14lowCD16- monocytes possessed limited ability for TNF-α production, and failed to suppress T-cell proliferation mediated by T-cell receptor signalling. However, these immature cells were highly proliferative and could differentiate into TNF-α producing, mature CD14highCD16+ monocytes. In the rat model of CPB, we further demonstrated that CPB induced migration of immature monocytes into the lungs, either from the bone marrow or from the spleen. Moreover, we confirmed the hypothesis that immature subsets could contribute to CPB-induced acute lung injury by giving rise to TNF-α producing descendants. CONCLUSIONS The immature CD14lowCD16- monocytes might contribute to blood-circuit contact-induced acute lung injury by generating TNF-α-producing, mature monocytes. New strategies based on monocyte manipulation could be a promising therapeutic approach for minimising CPB-related lung injury.
Collapse
Affiliation(s)
- Zhichen Xing
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junyan Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Xing Hao
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jinhong Wang
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chunjing Jiang
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yu Hao
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Hong Wang
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xueying Wu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Liwei Shen
- Department of Gastroenterology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xiaojun Dong
- Department of Gastroenterology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Institute of Digestive Disease, Shanghai, China
| | - Tong Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Guoli Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Jianping Zhang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Xiaotong Hou
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hui Zeng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| |
Collapse
|
47
|
Weber C, Shantsila E, Hristov M, Caligiuri G, Guzik T, Heine GH, Hoefer IE, Monaco C, Peter K, Rainger E, Siegbahn A, Steffens S, Wojta J, Lip GYH. Role and analysis of monocyte subsets in cardiovascular disease. Joint consensus document of the European Society of Cardiology (ESC) Working Groups "Atherosclerosis & Vascular Biology" and "Thrombosis". Thromb Haemost 2016; 116:626-37. [PMID: 27412877 DOI: 10.1160/th16-02-0091] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/02/2016] [Indexed: 12/21/2022]
Abstract
Monocytes as cells of the innate immunity are prominently involved in the development of atherosclerotic lesions. The heterogeneity of blood monocytes has widely been acknowledged by accumulating experimental and clinical data suggesting a differential, subset-specific contribution of the corresponding subpopulations to the pathology of cardiovascular and other diseases. This document re-evaluates current nomenclature and summarises key findings on monocyte subset biology to propose a consensus statement about phenotype, separation and quantification of the individual subsets.
Collapse
Affiliation(s)
- Christian Weber
- Dr. Christian Weber, LMU Munich - Cardiovascular Prevention, Pettenkoferstr. 9, 80336 Munich, Germany, Tel.: +49 89 4400 54350, Fax: +49 89 4400 54352, E-mail:
| | | | - Michael Hristov
- Dr. Michael Hristov, LMU Munich - Cardiovascular Prevention, Pettenkoferstr. 9, 80336 Munich, Germany, Tel.: +49 89 4400 54350, Fax: +49 89 4400 54352, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Westover AJ, Johnston KA, Buffington DA, Humes HD. An Immunomodulatory Device Improves Insulin Resistance in Obese Porcine Model of Metabolic Syndrome. J Diabetes Res 2016; 2016:3486727. [PMID: 27819007 PMCID: PMC5081446 DOI: 10.1155/2016/3486727] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/31/2016] [Indexed: 01/07/2023] Open
Abstract
Obesity is associated with tissue inflammation which is a crucial etiology of insulin resistance. This inflammation centers around circulating monocytes which form proinflammatory adipose tissue macrophages (ATM). Specific approaches targeting monocytes/ATM may improve insulin resistance without the adverse side effects of generalized immunosuppression. In this regard, a biomimetic membrane leukocyte processing device, called the selective cytopheretic device (SCD), was evaluated in an Ossabaw miniature swine model of insulin resistance with metabolic syndrome. Treatment with the SCD in this porcine model demonstrated a decline in circulating neutrophil activation parameters and monocyte counts. These changes were associated with improvements in insulin resistance as determined with intravenous glucose tolerance testing. These improvements were also reflected in lowering of homeostatic model assessment- (HOMA-) insulin resistant (IR) scores for up to 2 weeks after SCD therapy. These results allow for the planning of first-in-man studies in obese type 2 diabetic patients.
Collapse
Affiliation(s)
- Angela J. Westover
- Innovative BioTherapies, Inc., 650 Avis Drive, Suite 300, Ann Arbor, MI 48108, USA
| | - Kimberly A. Johnston
- Innovative BioTherapies, Inc., 650 Avis Drive, Suite 300, Ann Arbor, MI 48108, USA
| | | | - H. David Humes
- Innovative BioTherapies, Inc., 650 Avis Drive, Suite 300, Ann Arbor, MI 48108, USA
- Department of Internal Medicine, University of Michigan Medical School, 4520C MSRB I, SPC 5651, 1150 W. Medical Center Dr., Ann Arbor, MI 48109, USA
- *H. David Humes:
| |
Collapse
|
49
|
Li HJ, Zhai NC, Song HX, Yang Y, Cui A, Li TY, Tu ZK. The Role of Immune Cells in Chronic HBV Infection. J Clin Transl Hepatol 2015; 3:277-83. [PMID: 26807384 PMCID: PMC4721896 DOI: 10.14218/jcth.2015.00026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/20/2015] [Accepted: 10/28/2015] [Indexed: 12/20/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major cause of chronic liver diseases that may progress to liver cirrhosis and hepatocellular carcinoma. Host immune responses are important factors that determine whether HBV infection is cleared or persists. After infection, viral replication occurs inside hepatocytes, and the secretion of infectious virions can take place at high rates for decades. Consequently, HBV DNA and viral proteins, like HBV early antigen (HBeAg) and HBV surface antigen (HBsAg), can be easily detected in serum. Chronic infection with HBV is the result of an ineffective antiviral immune response towards the virus. In this review, we discuss the role of immune cells in chronic HBV infection.
Collapse
Affiliation(s)
- Hai-Jun Li
- Department of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Nai-Cui Zhai
- Department of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Hong-Xiao Song
- Department of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Yang Yang
- Department of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China
| | - An Cui
- Department of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tian-Yang Li
- Department of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Zheng-Kun Tu
- Department of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China
- Correspondence to: Zheng-Kun Tu, The First Hospital, Jilin University, Changchun 130061, Jilin, China. Tel: +86-0431-88783044, Fax: +86-0431-88783044, E-mail:
| |
Collapse
|
50
|
Sugimoto C, Hasegawa A, Saito Y, Fukuyo Y, Chiu KB, Cai Y, Breed MW, Mori K, Roy CJ, Lackner AA, Kim WK, Didier ES, Kuroda MJ. Differentiation Kinetics of Blood Monocytes and Dendritic Cells in Macaques: Insights to Understanding Human Myeloid Cell Development. THE JOURNAL OF IMMUNOLOGY 2015; 195:1774-81. [PMID: 26179903 DOI: 10.4049/jimmunol.1500522] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/16/2015] [Indexed: 12/23/2022]
Abstract
Monocyte and dendritic cell (DC) development was evaluated using in vivo BrdU pulse-chase analyses in rhesus macaques, and phenotype analyses of these cells in blood also were assessed by immunostaining and flow cytometry for comparisons among rhesus, cynomolgus, and pigtail macaques, as well as African green monkeys and humans. The nonhuman primate species and humans have three subsets of monocytes, CD14(+)CD16(-), CD14(+)CD16(+), and CD14(-)CD16(+) cells, which correspond to classical, intermediate, and nonclassical monocytes, respectively. In addition, there exist presently two subsets of DC, BDCA-1(+) myeloid DC and CD123(+) plasmacytoid DC, that were first confirmed in rhesus macaque blood. Following BrdU inoculation, labeled cells first appeared in CD14(+)CD16(-) monocytes, then in CD14(+)CD16(+) cells, and finally in CD14(-)CD16(+) cells, thus defining different stages of monocyte maturation. A fraction of the classical CD14(+)CD16(-) monocytes gradually expressed CD16(+) to become CD16(+)CD14(+) cells and subsequently matured into the nonclassical CD14(-)CD16(+) cell subset. The differentiation kinetics of BDCA-1(+) myeloid DC and CD123(+) plasmacytoid DC were distinct from the monocyte subsets, indicating differences in their myeloid cell origins. Results from studies utilizing nonhuman primates provide valuable information about the turnover, kinetics, and maturation of the different subsets of monocytes and DC using approaches that cannot readily be performed in humans and support further analyses to continue examining the unique myeloid cell origins that may be applied to address disease pathogenesis mechanisms and intervention strategies in humans.
Collapse
Affiliation(s)
- Chie Sugimoto
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Atsuhiko Hasegawa
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Yohei Saito
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Yayoi Fukuyo
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Kevin B Chiu
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Yanhui Cai
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Matthew W Breed
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Kazuyasu Mori
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Chad J Roy
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433; and
| | - Andrew A Lackner
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507
| | - Elizabeth S Didier
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433; and
| | - Marcelo J Kuroda
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA 70433;
| |
Collapse
|