1
|
Seizer L, Huber E, Schirmer M, Hilbert S, Wiest EM, Schubert C. Personalized therapy in rheumatoid arthritis (PETRA): a protocol for a randomized controlled trial to test the effect of a psychological intervention in rheumatoid arthritis. Trials 2023; 24:743. [PMID: 37986029 PMCID: PMC10659068 DOI: 10.1186/s13063-023-07707-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 10/06/2023] [Indexed: 11/22/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that primarily affects cartilage and bone. Psychological stress can both trigger disease exacerbation and result from disease activity. As standard pharmacological interventions alone have limited success in treating RA, a more comprehensive biopsychosocial approach to treatment has been recommended. In this prospective randomized controlled trial (RCT), a psychotherapeutically guided, group-based intervention program will be conducted with RA patients over a period of 9 months. This program combines a dynamic-interactional model with disorder-specific coping-oriented perspectives to improve patients' social, emotional, and problem-solving competencies as well as stress system functional status. The enrolment of 440 patients, randomly allocated to either an intervention (n = 220) or control group (n = 220), is planned. To evaluate the intervention effect, various indicators of RA disease activity, stress system activity, and psychological condition will be assessed through sets of standardized questionnaires and biochemical analyses of blood and saliva samples. Moreover, healthcare-related costs for each patient will be obtained using routine health insurance data. Outcome variables will be measured in all patients at regular intervals prior to intervention (baseline), during the 9-month intervention (five time points), and during a 9-month follow-up phase (three time points), allowing the comprehensive analysis of within- and between-subject effects, i.e. trajectories of the target variables in the intervention and control groups. In addition, to investigate the intervention effects on real-life stress system functioning in RA, 10 integrative single-case studies (n = 5 from the intervention group, n = 5 from the control group) will be conducted. In each study, once before and after the 9-month intervention, urine samples will be collected, and patients will fill out questionnaires for approximately 1 month at 12-h intervals. Moreover, weekly in-depth interviews will be conducted with patients to determine their previous week's emotionally positive and negative incidents. Using time series analysis, it is then possible to investigate whether and how stress system function in these RA patients has improved from the applied intervention. By using both an investigational macro- and microperspective, this project aims to evaluate a psychological intervention in the routine care of individuals with RA.Trial registration German Clinical Trials Register DRKS00028144. Registered on 1 March 2022.
Collapse
Affiliation(s)
- Lennart Seizer
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Tübingen, Tübingen, Germany
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Ellis Huber
- Professional Association of Preventologists, Berlin, Germany
| | - Miriam Schirmer
- Faculty of Human Sciences, University of Regensburg, Regensburg, Germany
| | - Sven Hilbert
- Faculty of Human Sciences, University of Regensburg, Regensburg, Germany
| | - Eva-Maria Wiest
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria
| | - Christian Schubert
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
2
|
Carvajal Gonczi CM, Hajiaghayi M, Gholizadeh F, Xavier Soares MA, Touma F, Lopez Naranjo C, Rios AJ, Pozzebon C, Daigneault T, Burchell-Reyes K, Darlington PJ. The β2-adrenergic receptor agonist terbutaline upregulates T helper-17 cells in a protein kinase A-dependent manner. Hum Immunol 2023; 84:515-524. [PMID: 37438188 DOI: 10.1016/j.humimm.2023.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/09/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND T helper 17 (Th17) cells produce IL-17A cytokine and can exacerbate autoimmune diseases and asthma. The β2 adrenergic receptor is a g protein-coupled receptor that induces cAMP second messenger pathways. We tested the hypothesis that terbutaline, a β2-adrenergic receptor-specific agonist, promotes IL-17 secretion by memory Th17 cells in a cAMP and PKA-dependent manner. METHODS Venous peripheral blood mononuclear cells (PBMC) from healthy human participants were activated with anti-CD3 and anti-CD28 antibodies. Secreted IL-17A was measured by enzyme linked immunosorbent assay, intracellular IL-17A, and RORγ were measured using flow cytometry, and RORC by qPCR. Memory CD3+CD4+CD45RA-CD45RO+ T cells were obtained by immunomagnetic negative selection and activated with tri-antibody complex CD3/CD28/CD2. Secreted IL-17A, intracellular IL-17A, RORC were measured, and phosphorylated-serine133-CREB was measured by western blotting memory Th cells. RESULTS Terbutaline increased IL-17A (p < 0.001), IL-17A+ cells (p < 0.05), and RORC in activated PBMC and memory Th cells. The PKA inhibitors H89 (p < 0.001) and Rp-cAMP (p < 0.01) abrogated the effects of terbutaline on IL-17A secretion in PBMC and memory T cells. Rolipram increased IL-17A (p < 0.01) to a similar extent as terbutaline. P-Ser133-CREB was increased by terbutaline (p < 0.05) in memory T cells. CONCLUSION Terbutaline augments memory Th17 cells in lymphocytes from healthy participants. This could exacerbate autoimmune diseases or asthma, in cases where Th17 cells are considered to be pro-inflammatory.
Collapse
Affiliation(s)
| | - Mehri Hajiaghayi
- Department of Biology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | - Fatemeh Gholizadeh
- Department of Biology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | | | - Fadi Touma
- Department of Biology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | | | - Amanda J Rios
- Department of Biology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | - Chelsea Pozzebon
- Department of Psychology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | - Tina Daigneault
- Department of Biology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | - Kelly Burchell-Reyes
- Department of Chemistry and Biochemistry, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | - Peter J Darlington
- Department of Biology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada; Department of Psychology, PERFORM Centre, Concordia University, Montreal, Quebec, Canada; Department of Health, Kinesiology & Applied Physiology, PERFORM Centre, Montreal, Quebec, Canada.
| |
Collapse
|
3
|
Nascimento DR, Balaniuc SLB, Palhares DB, Underwood A, Palhares MG, Alves F, Vieira FO, Souza-Fagundes EM, Giuliani LDR, Xavier PCN, Puerto HLD, Santos RAS, Milsted A, Brum JM, Silva IS, Martins AS. Rare and intractable fibrodysplasia ossificans progressiva shows different PBMC phenotype possibly modulated by ascorbic acid and propranolol treatment. Intractable Rare Dis Res 2021; 10:179-189. [PMID: 34466340 PMCID: PMC8397826 DOI: 10.5582/irdr.2021.01012] [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] [Academic Contribution Register] [Received: 01/15/2021] [Revised: 04/02/2021] [Accepted: 04/29/2021] [Indexed: 11/05/2022] Open
Abstract
Fibrodysplasia Ossificans Progressiva (FOP) is a rare congenital intractable disease associated with a mutation in ACVR1 gene, characterized by skeleton malformations. Ascorbic acid (AA) and propranolol (PP) in combination is reported to minimize flare-ups in patients. FOP leukocyte phenotype may possibly be modulated by AA and PP treatment. In this study, expression of 22 potential target genes was analyzed by RT-PCR in peripheral blood mononuclear cells culture (PBMC) from FOP patients and controls to determine effectiveness of the combination therapy. PBMC were treated with AA, PP and AA+PP combination. Basal expression of 12 of the 22 genes in FOP PBMC was statistically different from controls. ACVR1, ADCY2, ADCY9 and COL3 were downregulated while COL1 was upregulated. ADRB1, ADRB2, RUNX2, TNF-α and ACTB, were all overexpressed in FOP PBMC. In control, AA upregulated COL1, SVCT1, ACTB, AGTR2 and downregulated ADCY2. In FOP cells, AA upregulated ACVR1, BMP4, COL1, COL3, TNF-α, ADCY2, ADCY9, AGTR2 and MAS, while downregulated ADBR2, RUNX2, ADCY1, SVCT1 and ACTB. PP increased ADBR1 and decreased RUNX2, TNF-α, AGTR1, ACTB and CHRNA7 genes in treated control PBMC compared to untreated. PP upregulated ADBR1, ADBR2 and MAS, and downregulated TNF-α and ACTB in treated FOP PBMC versus untreated. AA+PP augmented ADRB1 and ADRB2 expressions in control PBMC. In FOP PBMC, AA+PP augmented ACVR1, COL1, COL3, ADBR1, AGTR2 and MAS expression and downregulated ADBR2, RUNX2, ACTB and MRGD. These data show distinct gene expression modulation in leukocytes from FOP patients when treated with AA and or PP.
Collapse
Affiliation(s)
| | | | | | - Adam Underwood
- Walsh University, Division of Mathematics and Sciences, North Canton, OH, USA
| | | | - Fabiana Alves
- UFMG/ Department of Physiology and Biophysics, Belo Horizonte, MG, Brazil
- Centro Universitário Metodista Izabela Hendrix- IMIH, Belo Horizonte, MG, Brazil
| | - Francisco Oliveira Vieira
- UFMG/ Department of Physiology and Biophysics, Belo Horizonte, MG, Brazil
- Centro Universitário Metodista Izabela Hendrix- IMIH, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | - Amy Milsted
- Walsh University, Division of Mathematics and Sciences, North Canton, OH, USA
| | - Jose Mauro Brum
- Procter & Gamble Health Care & Global Clinical Sciences, Mason, OH, USA
| | | | - Almir Sousa Martins
- UFMS/ Faculty of Medicine, Campo Grande, MS, Brazil
- UFMG/ Department of Physiology and Biophysics, Belo Horizonte, MG, Brazil
- Address correspondence to:Almir Sousa Martins, UFMG/ Department of Physiology and Biophysics, Av Antonio Carlos, 6627, A4-256, Belo Horizonte, MG, Brasil - 31.270-900. E-mail: ;
| |
Collapse
|
4
|
Markousis-Mavrogenis G, Tromp J, Ouwerkerk W, Ferreira JP, Anker SD, Cleland JG, Dickstein K, Filippatos G, Lang CC, Metra M, Samani NJ, de Boer RA, van Veldhuisen DJ, Voors AA, van der Meer P. Multimarker profiling identifies protective and harmful immune processes in heart failure: findings from BIOSTAT-CHF. Cardiovasc Res 2021; 118:1964-1977. [PMID: 34264317 PMCID: PMC9239579 DOI: 10.1093/cvr/cvab235] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/22/2021] [Revised: 06/16/2021] [Indexed: 11/18/2022] Open
Abstract
Aims The exploration of novel immunomodulatory interventions to improve outcome in heart
failure (HF) is hampered by the complexity/redundancies of inflammatory pathways, which
remain poorly understood. We thus aimed to investigate the associations between the
activation of diverse immune processes and outcomes in patients with HF. Methods and results We measured 355 biomarkers in 2022 patients with worsening HF and an independent
validation cohort (n = 1691) (BIOSTAT-CHF index and validation
cohorts), and classified them according to their functions into biological processes
based on the gene ontology classification. Principal component analyses were used to
extract weighted scores per process. We investigated the association of these processes
with all-cause mortality at 2-year follow-up. The contribution of each biomarker to the
weighted score(s) of the processes was used to identify potential therapeutic targets.
Mean age was 69 (±12.0) years and 537 (27%) patients were women. We identified 64 unique
overrepresented immune-related processes representing 188 of 355 biomarkers. Of these
processes, 19 were associated with all-cause mortality (10 positively and 9 negatively).
Increased activation of ‘T-cell costimulation’ and ‘response to
interferon-gamma/positive regulation of interferon-gamma production’ showed
the most consistent positive and negative associations with all-cause mortality,
respectively, after external validation. Within T-cell costimulation,
inducible costimulator ligand, CD28, CD70, and tumour necrosis factor superfamily
member-14 were identified as potential therapeutic targets. Conclusions We demonstrate the divergent protective and harmful effects of different immune
processes in HF and suggest novel therapeutic targets. These findings constitute a rich
knowledge base for informing future studies of inflammation in HF.
Collapse
Affiliation(s)
| | | | - Wouter Ouwerkerk
- Saw Swee Hock School of Public Health, National University of
Singapore, 12 Science Drive 2, #10-01, Singapore
117549, Singapore
- Department of Dermatology, Amsterdam UMC, University of Amsterdam,
Amsterdam Infection & Immunity Institute, De Boelelaan
1117, 1118, 1081 HV Amsterdam, The
Netherlands
| | - João Pedro Ferreira
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques, -
PlurithÕmatique 14-33, and Inserm U1116, CHRU, F-CRIN
INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
- Cardiovascular Research and Development Center, Department of Surgery and
Physiology, Faculty of Medicine of the University of Porto,
Porto, Portugal
| | - Stefan D Anker
- Division of Cardiology and Metabolism – Heart Failure, Cachexia &
Sarcopenia, Department of Cardiology (CVK), Berlin-Brandenburg Center for Regenerative
Therapies (BCRT), at Charité University Medicine, Charitépl.
1, 10117 Berlin, Germany
- Department of Cardiology and Pneumology, University Medicine Göttingen
(UMG), Robert-Koch-Straße 40, 37075 Göttingen,
Germany
- DZHK (German Center for Cardiovascular Research),
Potsdamer Str. 58 10785 Berlin, Germany
| | - John G Cleland
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing,
University of Glasgow, Glasgow G12 8QQ, UK
- National Heart & Lung Institute, Imperial College,
Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Kenneth Dickstein
- University of Bergen, Stavanger University Hospital,
Gerd-Ragna Bloch Thorsens gate 8, 4011 Stavanger, Norway
| | - Gerasimos Filippatos
- Heart Failure Unit, Department of Cardiology, National and Kapodistrian
University of Athens, School of Medicine, Athens University Hospital
Attikon, Rimini 1, Chaidari 124 62, Athens,
Greece
| | - Chim C Lang
- Division of Molecular & Clinical Medicine, University of
Dundee, Dundee DD1 9SY, UK
| | - Marco Metra
- Department of Medical and Surgical Specialties, Radiological Sciences and
Public Health, Institute of Cardiology, University of Brescia,
Piazza del Mercato, 15, 25121 Brescia BS, Italy
| | - Nilesh J Samani
- Division of Molecular & Clinical Medicine, University of
Dundee, Dundee DD1 9SY, UK
| | | | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University
of Groningen, Hanzeplein 1, 9713 GZ Groningen,
TheNetherlands
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University
of Groningen, Hanzeplein 1, 9713 GZ Groningen,
TheNetherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University
of Groningen, Hanzeplein 1, 9713 GZ Groningen,
TheNetherlands
| | | |
Collapse
|
5
|
Noah TK, Lee JB, Brown CA, Yamani A, Tomar S, Ganesan V, Newberry RD, Huffnagle GB, Divanovic S, Hogan SP. Thermoneutrality Alters Gastrointestinal Antigen Passage Patterning and Predisposes to Oral Antigen Sensitization in Mice. Front Immunol 2021; 12:636198. [PMID: 33841417 PMCID: PMC8034294 DOI: 10.3389/fimmu.2021.636198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2020] [Accepted: 03/05/2021] [Indexed: 01/02/2023] Open
Abstract
Food allergy is an emerging epidemic, and the underlying mechanisms are not well defined partly due to the lack of robust adjuvant free experimental models of dietary antigen sensitization. As housing mice at thermoneutrality (Tn) - the temperature of metabolic homeostasis (26-30°C) - has been shown to improve modeling various human diseases involved in inflammation, we tested the impact of Tn housing on an experimental model of food sensitization. Here we demonstrate that WT BALB/c mice housed under standard temperature (18-20°C, Ts) conditions translocated the luminal antigens in the small intestine (SI) across the epithelium via goblet cell antigen passages (GAPs). In contrast, food allergy sensitive Il4raF709 mice housed under standard temperature conditions translocated the luminal antigens in the SI across the epithelium via secretory antigen passages (SAPs). Activation of SI antigen passages and oral challenge of Il4raF709 mice with egg allergens at standard temperature predisposed Il4raF709 mice to develop an anaphylactic reaction. Housing Il4raF709 mice at Tn altered systemic type 2 cytokine, IL-4, and the landscape of SI antigen passage patterning (villus and crypt involvement). Activation of SI antigen passages and oral challenge of Il4raF709 mice with egg antigen under Tn conditions led to the robust induction of egg-specific IgE and development of food-induced mast cell activation and hypovolemic shock. Similarly, Tn housing of WT BALB/c mice altered the cellular patterning of SI antigen passage (GAPs to SAPs). Activation of SI antigen passages and the oral challenge of WT BALB/c mice with egg antigen led to systemic reactivity to egg and mast cell activation. Together these data demonstrate that Tn housing alters antigen passage cellular patterning and landscape, and concurrent oral exposure of egg antigens and SAP activation is sufficient to induce oral antigen sensitization.
Collapse
MESH Headings
- Administration, Oral
- Allergens/administration & dosage
- Allergens/immunology
- Allergens/metabolism
- Anaphylaxis/immunology
- Anaphylaxis/metabolism
- Anaphylaxis/microbiology
- Animals
- Disease Models, Animal
- Egg Hypersensitivity/immunology
- Egg Hypersensitivity/metabolism
- Egg Hypersensitivity/microbiology
- Egg Proteins/administration & dosage
- Egg Proteins/immunology
- Egg Proteins/metabolism
- Gastrointestinal Microbiome
- Goblet Cells/immunology
- Goblet Cells/metabolism
- Goblet Cells/microbiology
- Housing, Animal
- Intestine, Small/immunology
- Intestine, Small/metabolism
- Intestine, Small/microbiology
- Mast Cells/immunology
- Mast Cells/metabolism
- Mice, Inbred BALB C
- Mice, Knockout
- Permeability
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Temperature
- Mice
Collapse
Affiliation(s)
- Taeko K. Noah
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Jee-Boong Lee
- Division of Allergy and Immunology, Cincinnati Children’s Medical Center, Cincinnati, OH, United States
| | - Christopher A. Brown
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Amnah Yamani
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Sunil Tomar
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Varsha Ganesan
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Rodney D. Newberry
- Department of Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO, United States
| | - Gary B. Huffnagle
- Mary H Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children’s Medical Center, Cincinnati, OH, United States
- Center for Inflammation and Tolerance, Cincinnati Children’s Medical Center, Cincinnati, OH, United States
| | - Simon P. Hogan
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
- Mary H Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
6
|
Shifts of Immune Cell Populations Differ in Response to Different Effectors of Beige Remodeling of Adipose Tissue. iScience 2020; 23:101765. [PMID: 33294778 PMCID: PMC7683338 DOI: 10.1016/j.isci.2020.101765] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2020] [Revised: 09/09/2020] [Accepted: 10/30/2020] [Indexed: 11/21/2022] Open
Abstract
White adipose tissue (WAT) is a dynamic tissue, which responds to environmental stimuli and dietary cues by changing its morphology and metabolic capacity. The ability of WAT to undergo a beige remodeling has become an appealing strategy to combat obesity and its comorbidities. Here, by using single-cell RNA sequencing, we provide a comprehensive atlas of the cellular dynamics during beige remodeling. We reveal drastic changes both in the overall cellular composition and transcriptional states of individual cell subtypes between Adrb3- and cold-induced beiging. Moreover, we demonstrate that cold induces a myeloid to lymphoid shift of the immune compartment compared to Adrb3 activation. Further analysis showed that, Adrb3 stimulation leads to activation of the interferon/Stat1 pathways favoring infiltration of myeloid immune cells, while repression of this pathway by cold promotes lymphoid immune cell recruitment. These findings highlight that pharmacological mimetics may not provide the same beneficial effects as physiological stimuli.
Collapse
|
7
|
Almenara S, Lozano B, Gimenez P, Herrera I, Miralles C, Bellot P, Rodríguez M, Francés R, Gonzalez-Navajas JM, Pascual S, Zapater P. Functionality of beta-adrenergic receptors in patients with cirrhosis treated chronically with non-selective beta-blockers. Hepatol Int 2020; 14:858-868. [DOI: 10.1007/s12072-020-10083-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/13/2020] [Accepted: 08/17/2020] [Indexed: 12/25/2022]
|
8
|
Dimitrov S, Lange T, Gouttefangeas C, Jensen ATR, Szczepanski M, Lehnnolz J, Soekadar S, Rammensee HG, Born J, Besedovsky L. Gα s-coupled receptor signaling and sleep regulate integrin activation of human antigen-specific T cells. J Exp Med 2019; 216:517-526. [PMID: 30755455 PMCID: PMC6400544 DOI: 10.1084/jem.20181169] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/21/2018] [Revised: 12/03/2018] [Accepted: 01/11/2019] [Indexed: 12/26/2022] Open
Abstract
This study demonstrates a regulatory role of Gαs-coupled receptor agonists (catecholamines, prostaglandins, and adenosine) and sleep on integrin activation on T cells in humans. The findings point to a mechanism by which T cell responses are altered in several conditions characterized by aberrant levels of these substances. Efficient T cell responses require the firm adhesion of T cells to their targets, e.g., virus-infected cells, which depends on T cell receptor (TCR)–mediated activation of β2-integrins. Gαs-coupled receptor agonists are known to have immunosuppressive effects, but their impact on TCR-mediated integrin activation is unknown. Using multimers of peptide major histocompatibility complex molecules (pMHC) and of ICAM-1—the ligand of β2-integrins—we show that the Gαs-coupled receptor agonists isoproterenol, epinephrine, norepinephrine, prostaglandin (PG) E2, PGD2, and adenosine strongly inhibit integrin activation on human CMV- and EBV-specific CD8+ T cells in a dose-dependent manner. In contrast, sleep, a natural condition of low levels of Gαs-coupled receptor agonists, up-regulates integrin activation compared with nocturnal wakefulness, a mechanism possibly underlying some of the immune-supportive effects of sleep. The findings are also relevant for several pathologies associated with increased levels of Gαs-coupled receptor agonists (e.g., tumor growth, malaria, hypoxia, stress, and sleep disturbances).
Collapse
Affiliation(s)
- Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany .,German Center for Diabetes Research (DZD), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Tanja Lange
- Clinic for Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Anja T R Jensen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Szczepanski
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Jannik Lehnnolz
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Surjo Soekadar
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.,Clinical Neurotechnology Laboratory, Department of Psychiatry and Psychotherapy, Neuroscience Research Center, Charité - University Medicine Berlin, Berlin, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany.,Partner Site Tübingen, German Cancer Consortium, Tübingen, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Moreno-Villanueva M, Feiveson AH, Krieger S, Kay Brinda A, von Scheven G, Bürkle A, Crucian B, Wu H. Synergistic Effects of Weightlessness, Isoproterenol, and Radiation on DNA Damage Response and Cytokine Production in Immune Cells. Int J Mol Sci 2018; 19:ijms19113689. [PMID: 30469384 PMCID: PMC6275019 DOI: 10.3390/ijms19113689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/24/2018] [Revised: 11/10/2018] [Accepted: 11/11/2018] [Indexed: 12/15/2022] Open
Abstract
The implementation of rotating-wall vessels (RWVs) for studying the effect of lack of gravity has attracted attention, especially in the fields of stem cells, tissue regeneration, and cancer research. Immune cells incubated in RWVs exhibit several features of immunosuppression including impaired leukocyte proliferation, cytokine responses, and antibody production. Interestingly, stress hormones influence cellular immune pathways affected by microgravity, such as cell proliferation, apoptosis, DNA repair, and T cell activation. These pathways are crucial defense mechanisms that protect the cell from toxins, pathogens, and radiation. Despite the importance of the adrenergic receptor in regulating the immune system, the effect of microgravity on the adrenergic system has been poorly studied. Thus, we elected to investigate the synergistic effects of isoproterenol (a sympathomimetic drug), radiation, and microgravity in nonstimulated immune cells. Peripheral blood mononuclear cells were treated with the sympathomimetic drug isoproterenol, exposed to 0.8 or 2 Gy γ-radiation, and incubated in RWVs. Mixed model regression analyses showed significant synergistic effects on the expression of the β2-adrenergic receptor gene (ADRB2). Radiation alone increased ADRB2 expression, and cells incubated in microgravity had more DNA strand breaks than cells incubated in normal gravity. We observed radiation-induced cytokine production only in microgravity. Prior treatment with isoproterenol clearly prevents most of the microgravity-mediated effects. RWVs may be a useful tool to provide insight into novel regulatory pathways, providing benefit not only to astronauts but also to patients suffering from immune disorders or undergoing radiotherapy.
Collapse
Affiliation(s)
- Maria Moreno-Villanueva
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
- Molecular Toxicology Group, Department of Biology, Box 628, University of Konstanz, 78457 Konstanz, Germany.
| | - Alan H Feiveson
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
| | | | - AnneMarie Kay Brinda
- Department of Biomedical Engineering, University of Minnesota, 312 Church Street SE, Minneapolis, MN 55455, USA.
| | - Gudrun von Scheven
- Molecular Toxicology Group, Department of Biology, Box 628, University of Konstanz, 78457 Konstanz, Germany.
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, Box 628, University of Konstanz, 78457 Konstanz, Germany.
| | - Brian Crucian
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
| | - Honglu Wu
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
| |
Collapse
|
10
|
Fountoulaki K, Tsiodras S, Polyzogopoulou E, Olympios C, Parissis J. Beneficial Effects of Vaccination on Cardiovascular Events: Myocardial Infarction, Stroke, Heart Failure. Cardiology 2018; 141:98-106. [DOI: 10.1159/000493572] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/23/2018] [Accepted: 09/07/2018] [Indexed: 12/28/2022]
Abstract
Influenza and pneumococcal infections have been suggested to be potential risk factors for causing adverse cardiovascular events, especially in high-risk patients. Vaccination against respiratory infections in patients with established cardiovascular disease (CVD) could serve as a potential cost-effective intervention to improve their clinical outcomes and cardiac societies have encouraged it. Previous studies have shown that influenza vaccination reduce mortality, acute coronary syndromes and hospitalization in patients with coronary heart disease (CHD) and/or heart failure (HF). However, there is a paucity of randomized prospective clinical trials in the field of the pneumococcal vaccination, and additional higher-quality evidence is needed. Furthermore, questions around the role of vaccination in the primary prevention of CVD, the optimal dose and timing are largely unanswered. The pathophysiologic mechanism in which vaccination provides cardiovascular protection may be related to the modification of the immune-inflammatory model of atherogenesis. The present review summarizes the current evidence and understanding for vaccination against influenza and streptococcus pneumoniae in CHD, HF and stroke and highlights its beneficial effect in the reduction of adverse cardiovascular events.
Collapse
|
11
|
Isoproterenol-induced beta-2 adrenergic receptor activation negatively regulates interleukin-2 signaling. Biochem J 2018; 475:2907-2923. [PMID: 30120106 DOI: 10.1042/bcj20180503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022]
Abstract
Regulation of intracellular signaling pathways in lymphocytes is critical for cell homeostasis and immune response. Interleukin-2 (IL-2), a key regulator of lymphocytes, signals following receptor-ligand engagement and subsequent recruitment and activation of effector proteins including JAKs and STATs. Lymphocytes can also be regulated by the central nervous system through the β2 adrenergic receptor (β2AR) pathway which can affect cell trafficking, proliferation, differentiation, and cytokine production. The cross-talk between these two signaling pathways represents an important mechanism that has yet to be fully elucidated. The present study provides evidence for communication between the IL-2 receptor (IL-2R) and β2AR. Treatment of human lymphoid cell lines with the β2AR agonist isoproterenol (ISO) alone increased cAMP levels and mediated a stimulatory response by activating AKT and ERK to promote cell viability. Interestingly, ISO activation of β2AR also induced threonine phosphorylation of the IL-2Rβ. In contrast, ISO treatment prior to IL-2 stimulation produced an inhibitory signal that disrupted IL-2 induced activation of the JAK/STAT, MEK/ERK, and PI3K pathways by inhibiting the formation of the IL-2R beta-gamma chain complex, and subsequently cell proliferation. Moreover, γc-family cytokines-mediated STAT5 activation was also inhibited by ISO. These results suggest a molecular mechanism by which β2AR signaling can both stimulate and suppress lymphocyte responses and thus explain how certain therapeutic agents, such as vasodilators, may impact immune responsiveness.
Collapse
|
12
|
Salivary kynurenic acid response to psychological stress: inverse relationship to cortical glutamate in schizophrenia. Neuropsychopharmacology 2018; 43:1706-1711. [PMID: 29728648 PMCID: PMC6006286 DOI: 10.1038/s41386-018-0072-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/03/2018] [Revised: 04/04/2018] [Accepted: 04/09/2018] [Indexed: 12/22/2022]
Abstract
Frontal glutamatergic synapses are thought to be critical for adaptive, long-term stress responses. Prefrontal cortices, including the anterior cingulate cortex (ACC) contribute to stress perception and regulation, and are involved in top-down regulation of peripheral glucocorticoid and inflammatory responses to stress. Levels of kynurenic acid (KYNA) in saliva increase in response to psychological stress, and this stress-induced effect may be abnormal in people with schizophrenia. Here we test the hypothesis that ACC glutamatergic functioning may contribute to the stress-induced salivary KYNA response in schizophrenia. In 56 patients with schizophrenia and 58 healthy controls, our results confirm that levels of KYNA in saliva increase following psychological stress. The magnitude of the effect correlated negatively with proton magnetic resonance spectroscopy (MRS) glutamate + glutamine (r = -.31, p = .017) and glutamate (r = -0.27, p = .047) levels in the ACC in patients but not in the controls (all p ≥ .45). Although, a causal relationship cannot be ascertained in this cross-sectional study, these findings suggest a potentially meaningful link between central glutamate levels and kynurenine pathway response to stress in individuals with schizophrenia.
Collapse
|
13
|
The influence of biogenic amines on Th17-mediated immune response in multiple sclerosis. Mult Scler Relat Disord 2018; 21:19-23. [DOI: 10.1016/j.msard.2018.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/31/2017] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 12/13/2022]
|
14
|
Carvajal Gonczi CM, Tabatabaei Shafiei M, East A, Martire E, Maurice-Ventouris MHI, Darlington PJ. Reciprocal modulation of helper Th1 and Th17 cells by the β2-adrenergic receptor agonist drug terbutaline. FEBS J 2017; 284:3018-3028. [PMID: 28710773 DOI: 10.1111/febs.14166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/22/2016] [Revised: 06/02/2017] [Accepted: 07/12/2017] [Indexed: 01/01/2023]
Abstract
Catecholamine hormones are powerful regulators of the immune system produced by the sympathetic nervous system (SNS). They regulate the adaptive immune system by altering T-cell differentiation into T helper (Th) 1 and Th2 cell subsets, but the effect on Th17 cells is not known. Th17 cells, defined, in part, by chemokine receptor CCR6 and cytokine interleukin (IL)-17A, are crucial for mediating certain pathogen-specific responses and are linked with several autoimmune diseases. We demonstrated that a proportion of human Th17 cells express beta 2-adrenergic receptor (β2AR), a G protein-coupled receptor that responds to catecholamines. Activation of peripheral blood mononuclear cells, which were obtained from venous blood drawn from healthy volunteers, with anti-cluster of differentiation 3 (CD3) and anti-CD28 and with a β2-agonist drug, terbutaline (TERB), augmented IL-17A levels (P < 0.01) in the majority of samples. TERB reduced interferon gamma (IFNγ) indicating that IL-17A and IFNγ are reciprocally regulated. Similar reciprocal regulation was observed with dbcAMP. Proliferation of Th cells was monitored by carboxyfluorescein diacetate N-succinimidyl ester labeling and flow cytometry with antibody staining for CD3 and CD4. TERB increased proliferation by a small but significant margin (P < 0.001). Next, Th17 cells (CD4+ CXCR3- CCR6+ ) were purified using an immunomagnetic positive selection kit, which removes all other mononuclear cells. TERB increased IL-17A from purified Th17 cells, which argues that TERB acts directly on Th17 cells. Thus, hormone signals from the SNS maintain a balance of Th cells subtypes through the β2AR.
Collapse
Affiliation(s)
- Catalina M Carvajal Gonczi
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada
| | - Mahdieh Tabatabaei Shafiei
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada
| | - Ashley East
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Exercise Science, Concordia University, Montreal, QC, Canada
| | - Erika Martire
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada
| | - Meagane H I Maurice-Ventouris
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Peter J Darlington
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada.,Department of Exercise Science, Concordia University, Montreal, QC, Canada
| |
Collapse
|
15
|
Giles DA, Moreno-Fernandez ME, Stankiewicz TE, Graspeuntner S, Cappelletti M, Wu D, Mukherjee R, Chan CC, Lawson MJ, Klarquist J, Sünderhauf A, Softic S, Kahn CR, Stemmer K, Iwakura Y, Aronow BJ, Karns R, Steinbrecher KA, Karp CL, Sheridan R, Shanmukhappa SK, Reynaud D, Haslam DB, Sina C, Rupp J, Hogan SP, Divanovic S. Thermoneutral housing exacerbates nonalcoholic fatty liver disease in mice and allows for sex-independent disease modeling. Nat Med 2017; 23:829-838. [PMID: 28604704 PMCID: PMC5596511 DOI: 10.1038/nm.4346] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/06/2017] [Accepted: 04/22/2017] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a common prelude to cirrhosis and hepatocellular carcinoma, is the most common chronic liver disease worldwide. Defining the molecular mechanisms underlying the pathogenesis of NAFLD has been hampered by a lack of animal models that closely recapitulate the severe end of the disease spectrum in humans, including bridging hepatic fibrosis. Here we demonstrate that a novel experimental model employing thermoneutral housing, as opposed to standard housing, resulted in lower stress-driven production of corticosterone, augmented mouse proinflammatory immune responses and markedly exacerbated high-fat diet (HFD)-induced NAFLD pathogenesis. Disease exacerbation at thermoneutrality was conserved across multiple mouse strains and was associated with augmented intestinal permeability, an altered microbiome and activation of inflammatory pathways that are associated with the disease in humans. Depletion of Gram-negative microbiota, hematopoietic cell deletion of Toll-like receptor 4 (TLR4) and inactivation of the IL-17 axis resulted in altered immune responsiveness and protection from thermoneutral-housing-driven NAFLD amplification. Finally, female mice, typically resistant to HFD-induced obesity and NAFLD, develop full disease characteristics at thermoneutrality. Thus, thermoneutral housing provides a sex-independent model of exacerbated NAFLD in mice and represents a novel approach for interrogation of the cellular and molecular mechanisms underlying disease pathogenesis.
Collapse
Affiliation(s)
- Daniel A Giles
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Traci E Stankiewicz
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Monica Cappelletti
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David Wu
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rajib Mukherjee
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Calvin C Chan
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Matthew J Lawson
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jared Klarquist
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Annika Sünderhauf
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Samir Softic
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Bruce J Aronow
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rebekah Karns
- Division of Gastroenterology Hepatology and Nutrition, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kris A Steinbrecher
- Division of Gastroenterology Hepatology and Nutrition, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Rachel Sheridan
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shiva K Shanmukhappa
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David B Haslam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christian Sina
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Senad Divanovic
- Department of Pediatrics, Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
16
|
Shu J, Zhang F, Zhang L, Wei W. G protein coupled receptors signaling pathways implicate in inflammatory and immune response of rheumatoid arthritis. Inflamm Res 2016; 66:379-387. [DOI: 10.1007/s00011-016-1011-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2016] [Revised: 10/12/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
|
17
|
β2-Agonist clenbuterol hinders human monocyte differentiation into dendritic cells. Exp Cell Res 2015; 339:163-73. [PMID: 26524508 DOI: 10.1016/j.yexcr.2015.10.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/23/2015] [Revised: 10/27/2015] [Accepted: 10/29/2015] [Indexed: 12/20/2022]
Abstract
Clenbuterol (CLB) is a beta2-adrenergic agonist commonly used in asthma therapy, but is also a non-steroidal anabolic drug often abused in sport doping practices. Here we evaluated the in vitro impact of CLB on the physiology and function of human monocytes and dendritic cells (DCs), instrumental in the development of immune responses. We demonstrate that CLB inhibits the differentiation of monocytes into DCs and this effect is specific and dependent on β2-adrenergic receptor (AR) activation. We found that CLB treatment reduced the percentage of CD1a(+) immature DCs, while increasing the frequency of monocytes retaining CD14 surface expression. Moreover, CLB inhibited tumor necrosis factor-alpha (TNF-alpha) enhanced IL-(interleukin)-10 and IL-6 production. In contrast, CLB did not modulate the phenotypic and functional properties of monocytes and DCs, such as the surface expression of HLA-DR, CD83, CD80 and CD86 molecules, cytokine production, immunostimulatory activity and phagocytic activity. Moreover, we found that CLB did not modulate the activation of NF-kB in DCs. Moreover, we found that the differentiation of monocytes into DCs was associated with a significant decrease of β2-ARs mRNA expression. These results provide new insights on the effect of CLB on monocyte differentiation into DCs. Considering the frequent illegal use of CLB in doping, our work suggests that this drug is potentially harmful to immune responses decreasing the supply of DCs, thus subverting immune surveillance.
Collapse
|
18
|
Zalli A, Bosch JA, Goodyear O, Riddell N, McGettrick HM, Moss P, Wallace GR. Targeting ß2 adrenergic receptors regulate human T cell function directly and indirectly. Brain Behav Immun 2015; 45:211-8. [PMID: 25526818 DOI: 10.1016/j.bbi.2014.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/22/2014] [Revised: 11/30/2014] [Accepted: 12/01/2014] [Indexed: 01/28/2023] Open
Abstract
It is well-established that central nervous system activation affects peripheral blood mononuclear cell (PBMCs) function through the release of the catecholamines (Epi) and norepinephrine (NE), which act on ß2-adrenergic receptors (ß2AR). However, most studies have used non-specific stimulation of cells rather than antigen-specific responses. Likewise, few studies have parsed out the direct effects of ß2AR stimulation on T cells versus indirect effects via adrenergic stimulation of antigen presenting cells (APC). Here we report the effect of salmeterol (Sal), a selective ß2AR agonist, on IFN-γ(+) CD4 and IFN-γ(+) CD8 T cells following stimulation with Cytomegalovirus lysate (CMVL-strain AD169) or individual peptides spanning the entire region of the HCMV pp65 protein (pp65). Cells were also stimulated with Staphylococcal enterotoxin B. Additionally, we investigated the effect of Epi and Sal on cytotoxic cell killing of transfected target cells at the single cell level using the CD107a assay. The results show that Sal reduced the percentage of IFN-γ(+) CD4 and IFN-γ(+) CD8 T cells both when applied directly to isolated T cells, and indirectly via treatment of APC. These inhibitory effects were mediated via a ß2 adrenergic-dependent pathway and were stronger for CD8 as compared to CD4 T cells. Similarly, the results show that Sal suppressed cytotoxicity of both CD8 T and NK cells in vitro following stimulation with Chinese hamster ovary cell line transfected with MICA(*009) (T-CHO) and the human erythromyeloblastoid leukemic (K562) cell line. The inhibitory effect on cytotoxicity following stimulation with T-CHO was stronger in NK cells compared with CD8 T cells. Thus, targeting the ß2AR on lymphocytes and on APC leads to inhibition of inflammatory cytokine production and target cell killing. Moreover, there is a hierarchy of responses, with CD8 T cells and NK cells inhibited more effectively than CD4 T cells.
Collapse
Affiliation(s)
- A Zalli
- School of Sports and Exercise, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - J A Bosch
- Department of Psychology, University of Amsterdam, Amsterdam, The Netherlands.
| | - O Goodyear
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - N Riddell
- School of Sports and Exercise, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - H M McGettrick
- School of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - P Moss
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - G R Wallace
- School of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
19
|
Jenei-Lanzl Z, Grässel S, Pongratz G, Kees F, Miosge N, Angele P, Straub RH. Norepinephrine inhibition of mesenchymal stem cell and chondrogenic progenitor cell chondrogenesis and acceleration of chondrogenic hypertrophy. Arthritis Rheumatol 2014; 66:2472-81. [PMID: 24819295 DOI: 10.1002/art.38695] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2013] [Accepted: 05/02/2014] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Mesenchymal progenitor cell chondrogenesis is the biologic platform for the generation or regeneration of cartilage, but the external influence of the sympathetic nervous system on this process is not yet known. Sympathetic nerve fibers are present in articular tissue, and the sympathetic nervous system influences the musculoskeletal system by, for example, increasing osteoclastogenesis. This study was initiated to explore the role of the sympathetic neurotransmitter norepinephrine (NE) in mesenchymal stem cell (MSC)-dependent and cartilage progenitor cell (CPC)-dependent chondrogenesis. METHODS Using human MSCs or CPCs, chondrogenic differentiation was induced in the presence of NE, the specific β-adrenergic receptor (β-AR) agonist isoproterenol, and the specific β-AR antagonist nadolol. We studied sympathetic nerve fibers, tyrosine hydroxylase (TH) expression, catecholamine biosynthesis, and synovial fluid levels in human joints, as well as cartilage-specific matrix deposition during differentiation. RESULTS TH+ sympathetic nerve fibers were present in the synovial tissue, meniscus, and subchondral bone marrow. In addition, synovial fluid from patients with knee trauma demonstrated high concentrations of NE. During MSC or CPC chondrogenesis, β-AR were expressed. Chondrogenic aggregates treated with NE or isoproterenol synthesized lower amounts of type II collagen and glycosaminoglycans. NE and isoproterenol treatment dose-dependently increased the levels of cartilage hypertrophy markers (type X collagen and matrix metalloproteinase 13). Nadolol reversed the inhibition of chondrogenesis and the up-regulation of cartilage hypertrophy. CONCLUSION Our findings demonstrate NE-dependent inhibition of chondrogenesis and acceleration of hypertrophic differentiation. By inhibiting cartilage repair, these sympathetic influences can be important after joint trauma. These findings may be a basis for novel neurochondrogenic therapeutic options.
Collapse
|
20
|
Hilbert T, Bongartz J, Weisheit C, Knüfermann P, Baumgarten G, Hoeft A, Poth JM. Beta2-adrenoceptor stimulation suppresses TLR9-dependent IFNA1 secretion in human peripheral blood mononuclear cells. PLoS One 2013; 8:e65024. [PMID: 23724117 PMCID: PMC3665595 DOI: 10.1371/journal.pone.0065024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/22/2013] [Accepted: 04/19/2013] [Indexed: 11/24/2022] Open
Abstract
Introduction IFNA1 (interferon alpha) is a key cytokine regulating the activity of numerous immune cells. Plasmacytoid dendritic cells (pDCs) as natural interferon-producing cells play critical roles as sensors of pathogens and link innate to adaptive immunity. CpG motifs within DNA sequences activating toll-like receptor 9 (TLR9) are the main stimuli eliciting IFNA1 secretion from pDCs. Adrenergic substances are capable of differentially modulating the response from various immune cells. Hence, the aim of this study was to examine how adrenoceptor stimulation influences TLR9-induced IFNA1 secretion from human pDCs. Methods PBMCs generated from human whole blood and pDCs enriched from buffy coats were stimulated with LPS and CpG-ODN 2336 in the presence or absence of epinephrine and different adrenoceptor antagonists. Secretion of TNF and IFNA1 was measured by ELISA. Flow cytometry was used to determine efficacy of pDC enrichment and adrenoceptor expression of PBMC subsets. The influence of modified IFNA1 secretion on NK cell activity was evaluated using a colorimetric tumor cell lysis assay. Results TLR9-induced IFNA1 secretion as well as TLR4-induced TNF secretion from PBMCs was dose-dependently attenuated by coincubation with epinephrine. Combination with different specific adrenoceptor antagonists revealed that this effect was mediated by the adrenoceptor β2 (ADRB2). Since flow cytometric analysis could exclude the presence of ADRB2 on pDCs, highly enriched pDCs lacked any visible impact of adrenoceptor stimulation on TLR9-induced IFNA1 release. Combination of pDCs with PBMCs restored the effect, even when they were separated by a permeable membrane. Suppression of TLR9-mediated IFNA1 secretion from PBMCs by adrenoceptor stimulation reduced the lytic activity of NK cells on K562 tumor cells. Conclusion We provide insights into the underlying mechanisms of the interrelation between immune responses and pharmacological agents widely used in clinical practice. Our results have implications for the future treatment of human patients, in which the endogenous immune response plays a pivotal role, such as during viral infections, inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Tobias Hilbert
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Germany
| | - Josef Bongartz
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Germany
| | - Christina Weisheit
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Germany
| | - Pascal Knüfermann
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Germany
| | - Georg Baumgarten
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Germany
| | - Andreas Hoeft
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Germany
| | - Jens M. Poth
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Germany
- * E-mail:
| |
Collapse
|
21
|
Takayanagi Y, Osawa S, Ikuma M, Takagaki K, Zhang J, Hamaya Y, Yamada T, Sugimoto M, Furuta T, Miyajima H, Sugimoto K. Norepinephrine suppresses IFN-γ and TNF-α production by murine intestinal intraepithelial lymphocytes via the β₁ adrenoceptor. J Neuroimmunol 2012; 245:66-74. [PMID: 22398028 DOI: 10.1016/j.jneuroim.2012.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2011] [Revised: 01/16/2012] [Accepted: 02/10/2012] [Indexed: 11/28/2022]
Abstract
We examined whether norepinephrine (NE) had direct effects on cytokine production by murine intestinal intraepithelial lymphocytes (IELs), compared with splenocytes. CD3⁺ IELs and CD3⁺ splenocytes expressed α(1B), α(1D), α(2C), β₁, β₂, and β₃ adrenoceptors (ARs). NE significantly suppressed IFN-γ and TNF-α production by IELs and splenocytes ex vivo. The suppressive effects of NE in IELs were reversed by β₁ AR antagonist CGP-20712A, whereas those in splenocytes were reversed by β₂ AR antagonist ICI118,551. In IELs, β₁ AR agonist xamoterol mimicked the suppressive effects of NE. These results indicated NE regulates intestinal mucosal immune responses mediated by IELs via β₁ AR.
Collapse
Affiliation(s)
- Yasuhiro Takayanagi
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Fragala MS, Kraemer WJ, Mastro AM, Denegar CR, Volek JS, Häkkinen K, Anderson JM, Lee EC, Maresh CM. Leukocyte β2-adrenergic receptor expression in response to resistance exercise. Med Sci Sports Exerc 2011; 43:1422-32. [PMID: 21200338 DOI: 10.1249/mss.0b013e31820b88bc] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Epinephrine and norepinephrine mediate interactions between the neuroendocrine and the immune systems to alter immune cell activity. Although both systems respond to exercise stress, less is known about how they interact in response to such stress. The purpose of this investigation was to examine β2-adrenergic receptor (β2-ADR) expression on circulating leukocytes to an acute bout of resistance exercise in men and women. METHODS Resistance-trained men (n = 8; mean ± SD age = 24.63 ± 5.07 yr, body mass index = 26.09 ± 2.21 kg·m(-2)) and women (n = 7; age = 22.13 ± 3.09 yr, body mass index = 22.63 ± 2.03 kg·m(-2)) performed an acute resistance exercise protocol (six sets of five-repetition maximum heavy squats) and a control test (i.e., identical conditions with no exercise) in a balanced, randomized order. Using a within-subject design, β2-ADR expressions on circulating leukocytes were evaluated with flow cytometry, and plasma epinephrine and norepinephrine were evaluated with high-performance liquid chromatography. RESULTS Plasma epinephrine and norepinephrine increased during the exercise bout and returned to baseline during recovery. β2-ADR expression on monocytes was elevated in anticipation of the exercise protocol. β2-ADR expression on monocytes and granulocytes decreased during the exercise. β2-ADR expression on lymphocytes was elevated during the recovery time points. CONCLUSIONS In conclusion, β2-ADR expression on leukocyte subpopulations changes in response to acute heavy resistance exercise protocol. The present findings provide insights into the potential temporal interactions between the neuroendocrine and the immune systems in response to the physiological stress of acute heavy resistance exercise in men and women.
Collapse
Affiliation(s)
- Maren S Fragala
- Human Performance Laboratory, Department of Kinesiology, University of Connecticut, Storrs, CT 06269-1110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Koopman FA, Stoof SP, Straub RH, van Maanen MA, Vervoordeldonk MJ, Tak PP. Restoring the balance of the autonomic nervous system as an innovative approach to the treatment of rheumatoid arthritis. Mol Med 2011; 17:937-48. [PMID: 21607292 PMCID: PMC3188868 DOI: 10.2119/molmed.2011.00065] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/17/2011] [Accepted: 05/19/2011] [Indexed: 01/14/2023] Open
Abstract
The immunomodulatory effect of the autonomic nervous system has raised considerable interest over the last decades. Studying the influence on the immune system and the role in inflammation of the sympathetic as well as the parasympathetic nervous system not only will increase our understanding of the mechanism of disease, but also could lead to the identification of potential new therapeutic targets for chronic immune-mediated inflammatory diseases, such as rheumatoid arthritis (RA). An imbalanced autonomic nervous system, with a reduced parasympathetic and increased sympathetic tone, has been a consistent finding in RA patients. Studies in animal models of arthritis have shown that influencing the sympathetic (via α- and β-adrenergic receptors) and the parasympathetic (via the nicotinic acetylcholine receptor α7nAChR or by electrically stimulating the vagus nerve) nervous system can have a beneficial effect on inflammation markers and arthritis. The immunosuppressive effect of the parasympathetic nervous system appears less ambiguous than the immunomodulatory effect of the sympathetic nervous system, where activation can lead to increased or decreased inflammation depending on timing, doses and kind of adrenergic agent used. In this review we will discuss the current knowledge of the role of both the sympathetic (SNS) and parasympathetic nervous system (PNS) in inflammation with a special focus on the role in RA. In addition, potential antirheumatic strategies that could be developed by targeting these autonomic pathways are discussed.
Collapse
Affiliation(s)
- Frieda A Koopman
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, the Netherlands
| | - Susanne P Stoof
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, the Netherlands
- Arthrogen BV, Amsterdam, the Netherlands
| | - Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Marjolein A van Maanen
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, the Netherlands
| | - Margriet J Vervoordeldonk
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, the Netherlands
- Arthrogen BV, Amsterdam, the Netherlands
| | - Paul P Tak
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, the Netherlands
| |
Collapse
|
24
|
Chennaoui M, Sauvet F, Drogou C, Van Beers P, Langrume C, Guillard M, Gourby B, Bourrilhon C, Florence G, Gomez-Merino D. Effect of one night of sleep loss on changes in tumor necrosis factor alpha (TNF-α) levels in healthy men. Cytokine 2011; 56:318-24. [PMID: 21737301 DOI: 10.1016/j.cyto.2011.06.002] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/03/2011] [Revised: 05/23/2011] [Accepted: 06/02/2011] [Indexed: 01/12/2023]
Abstract
Total sleep deprivation in humans is associated with increased daytime sleepiness, decreased performance, elevations in inflammatory cytokines, and hormonal/metabolic disturbances. To assess the effects of 40 h of total sleep deprivation (TSD) under constant and well controlled conditions, on plasma levels of TNF-α and its receptor (TNFR1), interleukin-6 (IL-6), cortisol and C-reactive protein (CRP), sleepiness and performance, 12 healthy men (29±3 years) participated in a 5-days sleep deprivation experiment (two control nights followed by a night of sleep loss and one recovery night). Between 0800 and 2300 (i.e. between 25 and 40 h of sleep deprivation), a serial of blood sampling, multiple sleep latency, subjective levels of sleepiness and reaction time tests were completed before (day 2: D2) and after (day 4: D4) one night of sleep loss. We showed that an acute sleep deprivation (i.e. after 34 and 37 h of sleep deprivation) induced a significant increase in TNF-α (P<0.01), but there were no significant changes in TNFR1, IL-6, cortisol and CRP. In conclusion, our study in which constant and controlled experimental conditions were realized with healthy subjects and in absence of psychological or physical stressors, an acute total sleep deprivation (from 34 h) was sufficient to induce secretion of pro-inflammatory cytokine such as TNF-α, a marker more described in chronic sleep restriction or deprivation and as mediators of excessive sleepiness in humans in pathological conditions.
Collapse
Affiliation(s)
- M Chennaoui
- Institut de Recherche Biomédicale des Armées (IRBA), Département Environnements Opérationnels, Brétigny-sur-Orge, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sloan EK, Priceman SJ, Cox BF, Yu S, Pimentel MA, Tangkanangnukul V, Arevalo JMG, Morizono K, Karanikolas BDW, Wu L, Sood AK, Cole SW. The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res 2010; 70:7042-52. [PMID: 20823155 DOI: 10.1158/0008-5472.can-10-0522] [Citation(s) in RCA: 597] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
Metastasis to distant tissues is the chief driver of breast cancer-related mortality, but little is known about the systemic physiologic dynamics that regulate this process. To investigate the role of neuroendocrine activation in cancer progression, we used in vivo bioluminescence imaging to track the development of metastasis in an orthotopic mouse model of breast cancer. Stress-induced neuroendocrine activation had a negligible effect on growth of the primary tumor but induced a 30-fold increase in metastasis to distant tissues including the lymph nodes and lung. These effects were mediated by β-adrenergic signaling, which increased the infiltration of CD11b(+)F4/80(+) macrophages into primary tumor parenchyma and thereby induced a prometastatic gene expression signature accompanied by indications of M2 macrophage differentiation. Pharmacologic activation of β-adrenergic signaling induced similar effects, and treatment of stressed animals with the β-antagonist propranolol reversed the stress-induced macrophage infiltration and inhibited tumor spread to distant tissues. The effects of stress on distant metastasis were also inhibited by in vivo macrophage suppression using the CSF-1 receptor kinase inhibitor GW2580. These findings identify activation of the sympathetic nervous system as a novel neural regulator of breast cancer metastasis and suggest new strategies for antimetastatic therapies that target the β-adrenergic induction of prometastatic gene expression in primary breast cancers.
Collapse
Affiliation(s)
- Erica K Sloan
- UCLA Norman Cousins Center, Semel Institute for Neuroscience and Human Behavior, Medical Plaza 300, Room 3129, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Vardeny O, Moran JJM, Sweitzer NK, Johnson MR, Hayney MS. Decreased T-cell responses to influenza vaccination in patients with heart failure. Pharmacotherapy 2010; 30:10-6. [PMID: 20030468 DOI: 10.1592/phco.30.1.10] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2022]
Abstract
STUDY OBJECTIVE To determine whether T-cell immune responses to influenza vaccination in patients with chronic heart failure (CHF) are less vigorous than the responses of healthy control subjects. DESIGN Prospective, single-center study. SETTING University hospital and research laboratory. PARTICIPANTS Eighteen adults with stable CHF receiving optimal treatment and 16 healthy control subjects. INTERVENTION Participants were immunized with the 2006-2007 trivalent inactivated (killed) influenza vaccine during October-December of 2006. MEASUREMENTS AND MAIN RESULTS Blood samples were taken from the participants before and 2-4 weeks after vaccination to measure antibody titers, which were measured with a hemagglutination inhibition assay, then 3-4 months after vaccination to assess T-cell responses, measured by using the trans vivo delayed-type hypersensitivity method. As part of this method, which mimics physiologic conditions, peripheral blood mononuclear cells were isolated from the blood samples. The cells were mixed with influenza vaccine antigens A/H1N1, A/H3N2, and B type and injected into the footpads of SCID mice (mice with severe combined immunodeficiency), as their resulting swelling is an index of human T-cell sensitization. Median T-cell-mediated immune responses to A/H3N2 were less vigorous in patients with CHF than in control subjects (62.5 vs 87.5 microm, unadjusted p=0.031, age-adjusted p=0.006). Median responses to A/H1N1 were not significantly different between the groups (56.3 vs 75 microm, p=0.11). Median responses to B type were also similar between the groups (62.5 vs 75 microm, p=0.47). All participants mounted an antibody response to the influenza vaccine. CONCLUSION Patients with CHF had reduced T-cell responses to the influenza vaccine compared with healthy control subjects, as demonstrated by a lower response to A/H3N2, the newest antigen in the 2006-2007 vaccine. However, differences in T-cell immune responses to the A/H1N1 and B type strains were not found to be significant between the two groups, which suggests that patients with CHF can mount an appropriate response to vaccine antigens to which they have been previously exposed, but less so to new antigens. These findings suggest that patients with CHF may be at increased risk for influenza infection, and clinicians may want to investigate other or additional strategies for influenza vaccination.
Collapse
Affiliation(s)
- Orly Vardeny
- University of Wisconsin, Madison, 53705-2222, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Mollaret meningitis (MM) occurs mainly in females and is characterized by recurrent episodes of headache, transient neurological abnormalities, and the cerebrospinal fluid containing mononuclear cells. HSV-2 was usually identified as the causative agent. Recently, we found that recurrent headaches in non-HIV-infected subjects were due to acquired cerebral toxoplasmosis (CT). The aim of the study was therefore to focus on molecular pathomechanisms that may lead to reactivation of latent CT and manifest as MM. Literature data cited in this work were selected to illustrate that various factors may affect latent CNS Toxoplasma gondii infection/inflammation intensity and/or host defense mechanisms, i.e., the production of NO, cytokines, tryptophan degradation by indoleamine 2,3-dioxygenase, mechanisms mediated by an IFN-gamma responsive gene family, limiting the availability of intracellular iron to T. gondii, and production of reactive oxygen/nitrogen species, finally inducing choroid plexitis and/or vasculitis. Examples of triggers revealing MM and accompanying disturbances of IFN-gamma-mediated immune responses that control HSV-2 and T. gondii include: female predominance (female mice are more susceptible to T. gondii infection than males); HSV-2 infection (increased IFN-gamma, IL-12); metaraminol (increased plasma catecholamine levels, changes in cytokine expression favoring T(H)2 cells responses); probably cholesterol contained in debris from ruptured epidermoid cysts (decreased NO; increased TNF-alpha, IL-6, IL-8). These irregularities induced by the triggers may be responsible for reactivation of latent CT and development of MM. Thus, subjects with MM should have test(s) for T. gondii infection performed obligatorily.
Collapse
Affiliation(s)
- Joseph Prandota
- Pediatrics & Clinical Pharmacology, Department of Social Pediatrics, Faculty of Public Health, University Medical School, Wroclaw, Poland.
| |
Collapse
|
29
|
Irwin MR, Carrillo C, Olmstead R. Sleep loss activates cellular markers of inflammation: sex differences. Brain Behav Immun 2010; 24:54-7. [PMID: 19520155 PMCID: PMC2787978 DOI: 10.1016/j.bbi.2009.06.001] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/11/2009] [Revised: 05/23/2009] [Accepted: 06/02/2009] [Indexed: 12/20/2022] Open
Abstract
Sleep disturbance is associated with inflammation and related disorders including cardiovascular disease, arthritis, and diabetes mellitus. Given sex differences in the prevalence of inflammatory disorders with stronger associations in females, this study was undertaken to test the effects of sleep loss on cellular mechanisms that contribute to proinflammatory cytokine activity. In 26 healthy adults (11 females; 15 males), monocyte intracellular proinflammatory cytokine production was repeatedly assessed at 08:00, 12:00, 16:00, 20:00, and 23:00h during a baseline period and after partial sleep deprivation (awake from 23:00 to 3.00h). In the morning after a night of sleep loss, monocyte production of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) differentially changed between the two sexes. Whereas both females and males showed a marked increase in the lipopolysaccharide (LPS) - stimulated production of IL-6 and TNF-alpha in the morning immediately after PSD, production of these cytokines during the early- and late evening was increased in the females as compared to decreases in the males. Sleep loss induces a functional alteration of monocyte proinflammatory cytokine responses with females showing greater cellular immune activation as compared to changes in males. These results have implications for understanding the role of sleep disturbance in the differential risk profile for inflammatory disorders between the sexes.
Collapse
Affiliation(s)
- Michael R Irwin
- University of California, Los Angeles - Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA 90095-7076, USA.
| | | | | |
Collapse
|
30
|
Abstract
Pronounced circadian rhythms in numbers of circulating T cells reflect a systemic control of adaptive immunity whose mechanisms are obscure. Here, we show that circadian variations in T cell subpopulations in human blood are differentially regulated via release of cortisol and catecholamines. Within the CD4(+) and CD8(+) T cell subsets, naive cells show pronounced circadian rhythms with a daytime nadir, whereas (terminally differentiated) effector CD8(+) T cell counts peak during daytime. Naive T cells were negatively correlated with cortisol rhythms, decreased after low-dose cortisol infusion, and showed highest expression of CXCR4, which was up-regulated by cortisol. Effector CD8(+) T cells were positively correlated with epinephrine rhythms, increased after low-dose epinephrine infusion, and showed highest expression of beta-adrenergic and fractalkine receptors (CX3CR1). Daytime increases in cortisol via CXCR4 probably act to redistribute naive T cells to bone marrow, whereas daytime increases in catecholamines via beta-adrenoceptors and, possibly, a suppression of fractalkine signaling promote mobilization of effector CD8(+) T cells from the marginal pool. Thus, activation of the major stress hormones during daytime favor immediate effector defense but diminish capabilities for initiating adaptive immune responses.
Collapse
|
31
|
Rovensky J, Imrich R, Penesova A, Radikova Z, Scipova A, Vlcek M, Vigas M. Adrenomedullary response to hypoglycemia in first-degree relatives of patients with rheumatoid arthritis. Ann N Y Acad Sci 2009; 1148:552-5. [PMID: 19120156 DOI: 10.1196/annals.1410.070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
Our recent studies showed blunted adrenomedullary responses to insulin-induced hypoglycemia in premenopausal females with rheumatoid arthritis (RA) and systemic sclerosis, suggesting dysregulation of the adrenomedullary hormonal system (AMHS). Since no relationship has been found between degree of AMHS dysfunction and clinical or inflammatory parameters in those patients, we hypothesize the presence of an inherited perturbation of the AMHS. To test this hypothesis, we evaluated adrenomedullary responses to insulin-induced hypoglycemia (0.1 IU/kg) in premenopausal female subjects: 17 glucocorticoid-naïve RA patients, 15 healthy first-degree family members (FDR), and 18 age- and body mass index-matched healthy controls. Our results demonstrate that when compared to controls, RA patients had lower baseline epinephrine levels (P= 0.01) and lower area under response curve (AUC) levels of norepinephrine (P < 0.001) and epinephrine (P < 0.003). In contrast, FDR had lower (P= 0.001) AUC levels of norepinephrine compared to controls and higher (P= 0.033) AUC levels of epinephrine compared to RA patients. There were no significant differences in epinephrine response between FDR and controls. Although we found lower norepinephrine responses to hypoglycemia in FDR of RA patients, adrenomedullary responses to hypoglycemia does not appear to be altered to the degree found in RA patients.
Collapse
Affiliation(s)
- J Rovensky
- National Institute of Rheumatic Diseases, Piestany, Slovakia.
| | | | | | | | | | | | | |
Collapse
|
32
|
Vardeny O, Sweitzer NK, Detry MA, Moran JM, Johnson MR, Hayney MS. Decreased immune responses to influenza vaccination in patients with heart failure. J Card Fail 2008; 15:368-73. [PMID: 19398087 DOI: 10.1016/j.cardfail.2008.11.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/07/2008] [Revised: 10/21/2008] [Accepted: 11/14/2008] [Indexed: 11/26/2022]
Abstract
BACKGROUND Heart failure (HF) patients are at risk for influenza despite widespread vaccination. Both humoral (antibody) and cytotoxic T-lymphocyte (CTL) responses are important for protection. We explored antibody- and CTL-mediated responses to the influenza vaccine in HF patients compared with healthy controls. METHODS AND RESULTS We studied 29 HF patients (9 ischemic, 20 nonischemic) stable on HF therapies and 17 healthy controls. Participants had phlebotomy before and after influenza vaccination. Antibody production was measured in serum by hemagglutination inhibition assay and CTL responses (via interferon [IFN]-gamma and interleukin [IL]-10 production) were measured in isolated peripheral blood mononuclear cells with enzyme-linked immunosorbent assay. CTL responses demonstrated increased IL-10 production in HF patients after vaccination (P = .002), but similar IFN-gamma responses to healthy controls. All participants demonstrated antibody seroprotection; groups had similar rates of seroconversion (P = NS). Antibody-mediated response to the newest vaccine antigen, H3N2, was reduced in HF (P = .009). CONCLUSIONS Patients with HF had higher vaccine induced IL-10 concentrations, suggesting a different CTL phenotype for vaccine responses. HF patients did not mount as vigorous of an antibody immune response to the newest vaccine viral strain compared with healthy individuals. These data suggest that immunologic memory may be important for vaccine protection in HF patients.
Collapse
Affiliation(s)
- Orly Vardeny
- University of Wisconsin School of Pharmacy, Madison, Wisconsin 53705-2222, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Visvikis-Siest S, Marteau JB, Samara A, Berrahmoune H, Marie B, Pfister M. Peripheral blood mononuclear cells (PBMCs): a possible model for studying cardiovascular biology systems. Clin Chem Lab Med 2008; 45:1154-68. [PMID: 17663631 DOI: 10.1515/cclm.2007.255] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The inflammation system, alone or in relation to or interaction with other cardiovascular pathways, is suggested to be the central pathway in the development and progression of cardiovascular diseases. The aim of the present investigation was to propose a specific and informative model for exploring this hypothesis. METHODS In a biological system approach, we studied the expression of 182 candidate cardiovascular genes in peripheral blood mononuclear cells (PBMCs), cells that provide specific information on the inflammatory pathway. We explored their expression in 20 individuals with or without risk factors (obesity, hypertension) for cardiovascular disease. RESULTS We found that: 1) 166 among the 182 selected genes were expressed in at least one individual's PBMCs, some of them being detected for the first time in this tissue; 2) all pathways were represented by the majority of their genes selected; 3) genes were expressed at a level sufficient for further study of the inter-individual variations in their mRNA to determine their biological variation; and 4) 15 genes discriminated hypertensive from obese or controls. CONCLUSIONS The results of the present investigation support our proposal of a promising novel strategy based on PBMC transcriptomic studies to elucidate the complexity of the cardiovascular system in relation to inflammation. Preliminary data support the usefulness of the PBMC model in hypertension/inflammation research.
Collapse
|
34
|
Gosain A, Muthu K, Gamelli RL, DiPietro LA. Norepinephrine suppresses wound macrophage phagocytic efficiency through alpha- and beta-adrenoreceptor dependent pathways. Surgery 2007; 142:170-9. [PMID: 17689682 PMCID: PMC2430526 DOI: 10.1016/j.surg.2007.04.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2007] [Revised: 04/05/2007] [Accepted: 04/08/2007] [Indexed: 11/23/2022]
Abstract
BACKGROUND The systemic response to injury is characterized by massive release of norepinephrine (NE) into the circulation as a result of global sympathetic activation. We have recently demonstrated that NE modulates the recruitment of macrophages to the cutaneous wound. We hypothesized that NE suppresses wound macrophage phagocytic function through canonical adrenergic signaling pathways. METHODS Murine wound macrophages were harvested at 5 days after injury and treated with physiologic and pharmacologic dose norepinephrine. Phagocytosis of green fluorescent protein-labeled Escherichia coli was assayed by flow cytometry. The signaling pathways mediating NE modulation of wound macrophage phagocytosis were interrogated by pharmacologic manipulation of alpha- and beta-adrenoreceptors (ARs), intracellular cyclic adenosine monophosphate (cAMP), and protein kinase A (PKA). Tissue specificity was determined by comparison of wound macrophages to splenic macrophages. RESULTS Both physiologic and pharmacologic dose NE suppressed wound macrophage phagocytic efficiency. This effect was mediated by alpha- and beta-ARs in a dose-dependent fashion. Direct stimulation of cAMP-suppressed phagocytic efficiency and blockade of PKA signaling prevented NE-mediated suppression of phagocytic efficiency. Splenic macrophage phagocytic efficiency was less than that of wound macrophages and was not altered by NE. CONCLUSIONS NE has a profound immunosuppressive effect on wound macrophage function that is tissue specific and appears to be mediated through adrenergic receptors and their canonical downstream signaling pathway. Attenuation of post-injury immunosuppression represents another potential mechanism by which beta-AR blockade may reduce morbidity and mortality after severe injury.
Collapse
Affiliation(s)
- Ankush Gosain
- Burn and Shock Trauma Institute, Loyola University Medical Center, Maywood, IL, 60153
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University Medical Center, Maywood, IL, 60153
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153
| | - Kuzhali Muthu
- Burn and Shock Trauma Institute, Loyola University Medical Center, Maywood, IL, 60153
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153
| | - Richard L. Gamelli
- Burn and Shock Trauma Institute, Loyola University Medical Center, Maywood, IL, 60153
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153
| | - Luisa A. DiPietro
- Center for Wound Healing & Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, 60612
| |
Collapse
|
35
|
Boyle SH, Jackson WG, Suarez EC. Hostility, anger, and depression predict increases in C3 over a 10-year period. Brain Behav Immun 2007; 21:816-23. [PMID: 17321106 PMCID: PMC1995457 DOI: 10.1016/j.bbi.2007.01.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/30/2006] [Revised: 01/09/2007] [Accepted: 01/12/2007] [Indexed: 01/06/2023] Open
Abstract
We examined the relation of hostility, anger, and depression to 10-year changes in the third (C3), and fourth (C4) complement in 313, apparently healthy male participants enrolled in the Air Force Health Study (AFHS), a 20-year study designed to evaluate the health consequences of dioxin exposure. Hostility, depression, and anger were assessed using subscales from the Minnesota Multiphasic Personality Inventory (MMPI), which was administered in 1985. Given the high intercorrelations among these psychological scales, we used a principal component analysis to generate a composite score representing the linear combination of the hostility, anger, and depression scales. The dependent variables, C3 and C4 levels, were determined from samples collected in 1992, 1997, and 2002. Regression analyses controlling for age, race, alcohol use, body mass index, and cigarette use as well as onset of disease, and use of lipid lowering and blood pressure medications during follow-up revealed a significant timexcomposite score interaction for C3 complement (p<.0003), but not C4. Post-hoc analyses revealed that high composite scores were associated with larger 10-year increases in C3. These observations suggest that men who are hostile and are prone to experience frequent and intense feelings of anger, and depression show activation of the complement system, and specifically increases in C3, that may contribute to the development of coronary heart disease.
Collapse
Affiliation(s)
- Stephen H. Boyle
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center
| | | | - Edward C. Suarez
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center
| |
Collapse
|
36
|
O'Connor MF, Motivala SJ, Valladares EM, Olmstead R, Irwin MR. Sex differences in monocyte expression of IL-6: role of autonomic mechanisms. Am J Physiol Regul Integr Comp Physiol 2007; 293:R145-51. [PMID: 17428894 DOI: 10.1152/ajpregu.00752.2006] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022]
Abstract
Sex differences in the prevalence of inflammatory disorders exist, perhaps due to sex differences in cellular mechanisms that contribute to proinflammatory cytokine activity. This study analyzed sex differences of monocyte intracellular expression of IL-6 and its associations with reproductive hormones and autonomic mechanisms in 14 matched pairs of men and women ( n = 28). Monocyte intracellular IL-6 production was repeatedly assessed over two circadian periods. Sympathetic balance was estimated by heart rate variability and the ratio of power in the low-frequency (LF) to high-frequency (HF); vagal tone was indexed by the power of HF component. As compared to men, women showed greater monocyte expression of IL-6 across the circadian period. In addition, women showed lower sympathetic balance (LF/HF ratio), and greater levels of vagal tone (HF power). In women, but not men, sympathovagal balance was negatively associated with monocyte IL-6 expression, whereas vagal tone was positively associated with production of this cytokine. Levels of reproductive hormones were not related to monocyte IL-6 expression. The marked increase in monocyte expression of interleukin-6 in women has implications for understanding sex differences in risk of inflammatory disorders. Additionally, these data suggest that sex differences in sympathovagal balance or vagal tone may be a pathway to explain sex differences in IL-6 expression. Interventions that target autonomic mechanisms might constitute new strategies to constrain IL-6 production with impacts on inflammatory disease risk in women.
Collapse
Affiliation(s)
- Mary-Frances O'Connor
- Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience and Human Behavior, University of California-Los Angeles, 300 Medical Plaza, Los Angeles, CA 90095-7076, USA.
| | | | | | | | | |
Collapse
|
37
|
Stob NR, Seals DR, Jørgen J, van Baak MA, Steig AJ, Lindstrom RC, Bikman BT, Bell C. Increased thermogenic responsiveness to intravenous beta-adrenergic stimulation in habitually exercising humans is not related to skeletal muscle beta2-adrenergic receptor density. Exp Physiol 2007; 92:823-30. [PMID: 17573413 DOI: 10.1113/expphysiol.2007.038174] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/08/2022]
Abstract
Habitually exercising adults demonstrate greater thermogenic responsiveness to beta-adrenergic receptor (beta-AR) stimulation compared with their sedentary peers, but the molecular mechanisms involved are unknown. To determine the possible role of increased beta-AR density, we studied 32 healthy adults: 17 habitual aerobic exercisers (age 45 +/- 5 years, 11 males) and 15 sedentary (49 +/- 5 years, 7 males). Maximal oxygen uptake (43.7 +/- 2.5 versus 31.6 +/- 2.9 ml kg(-1) min(-1), P = 0.002, mean +/- S.E.M.) and vastus lateralis muscle maximal citrate synthase activity (1.70 +/- 0.36 versus 0.58 +/- 0.11 micromol min(-1) g(-1), P = 0.008) were higher in the habitually exercising subjects. Resting energy expenditure (EE) adjusted for fat-free mass (FFM) was similar in the habitually exercising (5903 +/- 280 kJ day(-1)) and sedentary adults (6054 +/- 289 kJ day(-1), P = 0.43). The percentage increase in EE (DeltaEE%; indirect calorimetry, ventilated hood) above resting EE in response to beta-AR stimulation (intravenous isoproterenol at 6, 12 and 24 ng (kg FFM)(-1) min(-1)) was greater (7.1 +/- 1.2, 13.7 +/- 1.0, 20.7 +/- 1.3 versus 5.9 +/- 0.9, 9.9 +/- 1.4, 15.9 +/- 1.70%, respectively, P = 0.04), and the dose of isoproterenol required to increase EE by 10% above resting EE was lower (8.2 +/- 1.5 versus 17.1 +/- 4.1 ng (kg FFM)(-1) min(-1), P = 0.03) in the habitually exercising adults. In contrast, vastus lateralis muscle beta(2)-AR density was similar in the habitually exercising and sedentary subjects (7.46 +/- 0.29 versus 7.44 +/- 0.60 fmol (mg dry weight muscle)(-1), P = 0.98), and was not related to DeltaEE% (r = 0.02, P = 0.94) or to the isoproterenol dose required to increase EE by 10% above resting EE (r = -0.06, P = 0.76). These findings indicate that increased beta(2)-AR density is not a mechanism contributing to the greater thermogenic responsiveness to beta-AR stimulation in adult humans who regularly perform aerobic exercise.
Collapse
Affiliation(s)
- Nicole R Stob
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO 80309-0354, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
de Coupade C, Brown AS, Dazin PF, Levine JD, Green PG. beta(2)-Adrenergic receptor-dependent sexual dimorphism for murine leukocyte migration. J Neuroimmunol 2007; 186:54-62. [PMID: 17442405 PMCID: PMC1994158 DOI: 10.1016/j.jneuroim.2007.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2006] [Revised: 02/15/2007] [Accepted: 02/28/2007] [Indexed: 01/03/2023]
Abstract
In wild-type FVB mice, leukocyte recruitment to lipopolysaccharide was sexually dimorphic, with a greater number of leukocytes recruited in females. In male beta(2)-adrenergic receptor knock out mice (bred on a congenic FVB background) the number of leukocytes recruited was increased approximately 4-fold, while in females there was no change, eliminating sexual dimorphism in leukocyte migration. While there were significantly fewer recruited CD62L(+) and CD11a(+) leukocytes in wild-type males, only in male beta-adrenergic receptor knock out mice was there an increase in the number of recruited CD11a(+) leukocytes, again eliminating sexual dimorphism. Thus, leukocyte migration and CD11a(+) adhesion molecule expression in male, but not in female, leukocytes is beta-adrenergic receptor-dependent. Our findings provide support for a role of beta(2)-adrenergic receptor mechanisms in the inflammatory response, and suggest that beta(2)-adrenergic receptor on male leukocytes contributes to sexual dimorphism in the effect of stress on inflammatory diseases.
Collapse
Affiliation(s)
- Catherine de Coupade
- Department of Medicine and Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA 94143
| | - Adrienne S. Brown
- Department of Medicine and Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA 94143
| | - Paul F. Dazin
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jon D. Levine
- Department of Medicine and Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA 94143
| | - Paul G. Green
- *Corresponding author: Department of Medicine and Oral and Maxillofacial Surgery, 521 Parnassus Avenue, UCSF, San Francisco, CA 94143, USA. Tel: +1 415 476 4902; Fax: 415-476-6305, E-mail:
| |
Collapse
|