1
|
Brod SA. Anti-Inflammatory Agents: An Approach to Prevent Cognitive Decline in Alzheimer's Disease. J Alzheimers Dis 2021; 85:457-472. [PMID: 34842189 DOI: 10.3233/jad-215125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Systemic inflammation is an organism's response to an assault by the non-self. However, that inflammation may predispose humans to illnesses targeted to organs, including Alzheimer's disease (AD). Lesions in AD have pro-inflammatory cytokines and activated microglial/monocyte/macrophage cells. Up to this point, clinical trials using anti-amyloid monoclonal antibodies have not shown success. Maybe it is time to look elsewhere by combating inflammation. Neuroinflammation with CNS cellular activation and excessive expression of immune cytokines is suspected as the "principal culprit" in the higher risk for sporadic AD. Microglia, the resident immune cell of the CNS, perivascular myeloid cells, and activated macrophages produce IL-1, IL-6 at higher levels in patients with AD. Anti-inflammatory measures that target cellular/cytokine-mediated damage provide a rational therapeutic strategy. We propose a clinical trial using oral type 1 IFNs to act as such an agent; one that decreases IL-1 and IL-6 secretion by activating lamina propria lymphocytes in the gut associated lymphoid tissue with subsequent migration to the brain undergoing inflammatory responses. A clinical trial would be double-blind, parallel 1-year clinical trial randomized 1 : 1 oral active type 1 IFN versus best medical therapy to determine whether ingested type I IFN would decrease the rate of cognitive decline in mild cognitive impairment or mild AD. Using cognitive psychometrics, imaging, and fluid biomarkers (MxA for effective type I IFN activity beyond the gut), we can determine if oral type I IFN can prevent cognitive decline in AD.
Collapse
Affiliation(s)
- Staley A Brod
- Department of Neurology, Medical College of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
2
|
Zhang X, Wang S, Zhu Y, Zhang M, Zhao Y, Yan Z, Wang Q, Li X. Double-edged effects of interferons on the regulation of cancer-immunity cycle. Oncoimmunology 2021; 10:1929005. [PMID: 34262796 PMCID: PMC8253121 DOI: 10.1080/2162402x.2021.1929005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Interferons (IFNs) are a large family of pleiotropic cytokines that regulate both innate and adaptive immunity and show anti-cancer effects in various cancer types. Moreover, it was revealed that IFN signaling plays critical roles in the success of cancer therapy strategies, thereby enhancing their therapeutic effects. However, IFNs have minimal or even adverse effects on cancer eradication, and mediate cancer immune escape in some instances. Thus, IFNs have a double-edged effect on the cancer immune response. Recent studies suggest that IFNs regulate each step of the cancer immunity-cycle, consisting of cancer antigen release, presentation of antigens and activation of T cells, trafficking and infiltration of effector T cells into the tumor microenvironment, and recognition and killing of cancer cells, which contributes to our understanding of the mechanisms of IFNs in regulating cancer immunity. In this review, we focus on IFNs and cancer immunity and elaborate on the roles of IFNs in regulating the cancer-immunity cycle.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Stomatology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Pathology, Harbin Medical University, Harbin, China
| | - Song Wang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yuanyuan Zhu
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Minghui Zhang
- Department of Oncology, Chifeng City Hospital, Chifeng, China
| | - Yan Zhao
- Department of Oncology, Chifeng City Hospital, Chifeng, China
| | - Zhengbin Yan
- Department of Stomatology, the PeopIe's Hospital of Longhua, Shenzhen, China
| | - Qiuxu Wang
- Department of Stomatology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Stomatology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xiaobo Li
- Department of Stomatology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of Pathology, Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Yoshimura S, Thome R, Konno S, Mari ER, Rasouli J, Hwang D, Boehm A, Li Y, Zhang GX, Ciric B, Rostami A. IL-9 Controls Central Nervous System Autoimmunity by Suppressing GM-CSF Production. THE JOURNAL OF IMMUNOLOGY 2019; 204:531-539. [PMID: 31852750 DOI: 10.4049/jimmunol.1801113] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/18/2019] [Indexed: 01/26/2023]
Abstract
Multiple sclerosis and experimental autoimmune encephalomyelitis (EAE) are inflammatory diseases of the CNS in which Th17 cells play a major role in the disease pathogenesis. Th17 cells that secrete GM-CSF are pathogenic and drive inflammation of the CNS. IL-9 is a cytokine with pleiotropic functions, and it has been suggested that it controls the pathogenic inflammation mediated by Th17 cells, and IL-9R-/- mice develop more severe EAE compared with wild-type counterparts. However, the underlying mechanism by which IL-9 suppresses EAE has not been clearly defined. In this study, we investigated how IL-9 modulates EAE development. By using mice knockout for IL-9R, we show that more severe EAE in IL-9R-/- mice correlates with increased numbers of GM-CSF+ CD4+ T cells and inflammatory dendritic cells (DCs) in the CNS. Furthermore, DCs from IL-9R-/- mice induced more GM-CSF production by T cells and exacerbated EAE upon adoptive transfer than did wild-type DCs. Our results suggest that IL-9 reduces autoimmune neuroinflammation by suppressing GM-CSF production by CD4+ T cells through the modulation of DCs.
Collapse
Affiliation(s)
- Satoshi Yoshimura
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Rodolfo Thome
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Shingo Konno
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Elisabeth R Mari
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Javad Rasouli
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Daniel Hwang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Alexandra Boehm
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Yanhua Li
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Guang-Xian Zhang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Bogoljub Ciric
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Abdolmohamad Rostami
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
4
|
Adriani M, Nytrova P, Mbogning C, Hässler S, Medek K, Jensen PEH, Creeke P, Warnke C, Ingenhoven K, Hemmer B, Sievers C, Lindberg Gasser RL, Fissolo N, Deisenhammer F, Bocskei Z, Mikol V, Fogdell-Hahn A, Kubala Havrdova E, Broët P, Dönnes P, Mauri C, Jury EC. Monocyte NOTCH2 expression predicts IFN-β immunogenicity in multiple sclerosis patients. JCI Insight 2018; 3:99274. [PMID: 29875313 DOI: 10.1172/jci.insight.99274] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/24/2018] [Indexed: 01/25/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by CNS inflammation leading to demyelination and axonal damage. IFN-β is an established treatment for MS; however, up to 30% of IFN-β-treated MS patients develop neutralizing antidrug antibodies (nADA), leading to reduced drug bioactivity and efficacy. Mechanisms driving antidrug immunogenicity remain uncertain, and reliable biomarkers to predict immunogenicity development are lacking. Using high-throughput flow cytometry, NOTCH2 expression on CD14+ monocytes and increased frequency of proinflammatory monocyte subsets were identified as baseline predictors of nADA development in MS patients treated with IFN-β. The association of this monocyte profile with nADA development was validated in 2 independent cross-sectional MS patient cohorts and a prospective cohort followed before and after IFN-β administration. Reduced monocyte NOTCH2 expression in nADA+ MS patients was associated with NOTCH2 activation measured by increased expression of Notch-responsive genes, polarization of monocytes toward a nonclassical phenotype, and increased proinflammatory IL-6 production. NOTCH2 activation was T cell dependent and was only triggered in the presence of serum from nADA+ patients. Thus, nADA development was driven by a proinflammatory environment that triggered activation of the NOTCH2 signaling pathway prior to first IFN-β administration.
Collapse
Affiliation(s)
- Marsilio Adriani
- Department of Rheumatology, University College Hospital, London, United Kingdom
| | - Petra Nytrova
- Department of Neurology and Center for Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Cyprien Mbogning
- CESP, Fac. De Médecine-Univ. Paris-Sud, Fac. De Médecine-UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
| | - Signe Hässler
- CESP, Fac. De Médecine-Univ. Paris-Sud, Fac. De Médecine-UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
| | - Karel Medek
- Department of Neurology and Center for Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Poul Erik H Jensen
- Neuroimmunology Laboratory, DMSC, Department of Neurology, Rigshospitalet, Region H, Copenhagen, Denmark
| | - Paul Creeke
- Neuroimmunology Unit, Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Clemens Warnke
- Department of Neurology, Medical Faculty, Research Group for Clinical and Experimental Neuroimmunology, Heinrich-Heine-University, Düsseldorf, Germany.,University Hospital Koeln, Deptartment of Neurology, Koeln, Germany
| | - Kathleen Ingenhoven
- Department of Neurology, Medical Faculty, Research Group for Clinical and Experimental Neuroimmunology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Bernhard Hemmer
- Klinikum rechts der Isar, Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Claudia Sievers
- Laboratory of Clinical Neuroimmunology, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Raija Lp Lindberg Gasser
- Laboratory of Clinical Neuroimmunology, Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Nicolas Fissolo
- Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Florian Deisenhammer
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Zsolt Bocskei
- Translational Sciences Unit, Sanofi R&D, 91385 Chilly-Mazarin, Paris, France
| | - Vincent Mikol
- Translational Sciences Unit, Sanofi R&D, 91385 Chilly-Mazarin, Paris, France
| | - Anna Fogdell-Hahn
- Karolinska Institutet, Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska University Hospital, Sweden
| | - Eva Kubala Havrdova
- Department of Neurology and Center for Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Philippe Broët
- CESP, Fac. De Médecine-Univ. Paris-Sud, Fac. De Médecine-UVSQ, INSERM, Université Paris-Saclay, Villejuif, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Paul Brousse, Villejuif, France
| | | | - Claudia Mauri
- Department of Rheumatology, University College Hospital, London, United Kingdom
| | - Elizabeth C Jury
- Department of Rheumatology, University College Hospital, London, United Kingdom
| | | |
Collapse
|
5
|
Ashtari F, Toghianifar N, Zarkesh-Esfahani SH, Mansourian M. Short-term effect of high-dose vitamin D on the level of interleukin 10 in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled clinical trial. Neuroimmunomodulation 2015; 22:400-4. [PMID: 26401986 DOI: 10.1159/000439278] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/06/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system. Vitamin D has been related to the prevention of MS and to modulating its course. Recent studies have shown the safety of high-dose vitamin D in MS. OBJECTIVE This study compared the effects of high-dose vitamin D on interleukin 10 (IL-10) levels in MS patients in a double-blind, randomized clinical trial. METHODS Ninety-four patients with relapsing remitting MS (RRMS) were randomized into a treatment and a placebo group. Both groups received conventional MS treatment. The intervention group received 50,000 IU of vitamin D every 5 days for 3 months. IL-10 was measured at baseline and after 3 months. RESULTS Serum levels of IL-10 were (median ± IQR): 12.58 ± 11.97 and 10.97 ± 9.97 pg/ml in the intervention and placebo groups, respectively, at baseline (p = 0.161); after 3 months, these levels were 13.76 ± 18.95 and 11.31 ± 19.63 pg/ml, respectively (p = 0.158). The IL-10 level increased significantly after receiving high-dose vitamin D for 3 months (β = 0.737, p = 0.015 and R2 = 0.91). CONCLUSION IL-10 levels increased significantly in RRMS patients after taking high-dose vitamin D3 for 3 months. High-dose vitamin D might be useful in promoting an anti-inflammatory state in RRMS patients.
Collapse
Affiliation(s)
- Fereshteh Ashtari
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | | |
Collapse
|
6
|
Patti F, Nicoletti A, Pappalardo A, Castiglione A, Lo Fermo S, Messina S, D'Amico E, Cimino V, Zappia M. Frequency and severity of headache is worsened by Interferon-β therapy in patients with multiple sclerosis. Acta Neurol Scand 2012; 125:91-5. [PMID: 21649611 DOI: 10.1111/j.1600-0404.2011.01532.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The relationship between multiple sclerosis (MS) and headache (HA) is not well known. It was reported that interferon-beta (IFNβ) could induce or worsen HA. OBJECTIVE To evaluate the impact of IFNβ treatment on HA and the relationship between HA and the various commercial preparations of IFNβ in mildly disabled patients with MS. METHODS A specific questionnaire was administered to 357 relapsing-remitting MS patients. Characteristics of HAs were considered, including the temporal relationships with IFNβ administration. RESULTS One hundred and seventeen patients were treated with weekly intramuscular injections of interferon IFNβ-1a (Avonex(®)), 84 with subcutaneous injections of IFNβ-1b (Betaferon(®)) every other day, 48 and 108 with three times weekly subcutaneous injections of IFNβ-1a (Rebif(®)) 22 mcg or IFNβ-1a (Rebif(®)) 44 mcg, respectively. Three hundred and fourteen patients were affected by HA, and among them, 219 patients suffered of pre-existing HA. In this latter group, 121 subjects (55%) noted a worsening of their HA after starting IFNβ therapy; this was more frequently reported by patients treated with Avonex(®) and Rebif(®) 44. Ninety-five patients experienced new HA. CONCLUSION IFNβ treatment could worsen HA in patients with pre-existing HA or cause the appearance of new HA. Among different IFNβ preparations, Rebif(®) 44 and Avonex(®) seemed to be more cephalalgic than the other drugs.
Collapse
Affiliation(s)
- F Patti
- Department of Neurosciences, University of Catania, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Disialogangliosides and TNFα alter gene expression for cytokines and chemokines in primary brain cell cultures. Neurochem Res 2011; 37:214-22. [PMID: 21964763 DOI: 10.1007/s11064-011-0587-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 08/24/2011] [Accepted: 08/29/2011] [Indexed: 10/17/2022]
Abstract
Gangliosides have long been implicated in multiple pathologies affecting the central nervous system. Empirical studies have suggested the possibility that gangliosides, particularly GD3, work in tandem with pro-inflammatory cytokines, especially tumor necrosis factor alpha (TNFα), to initiate or facilitate cell death in the CNS. As a step toward unraveling the metabolic pathways activated in the pathogenesis of brain cell death, we have surveyed gene expression for a host of cytokines and chemokines in primary brain cell cultures exposed to GD3, GD1b, and TNFα for 24 h. An initial screen of 98 genes on a focused mini-array revealed the expression of at least 28 genes related to cell growth, death, or inflammation in our system of mixed cells cultured from neonatal rat brains. Clear evidence of a differential response to the gangliosides or TNFα was seen in 12 genes. Quantitative PCR was used to validate the response of six of these genes. We found that both GD3 and GD1b, but not TNFα, up-regulated expression of macrophage inflammatory protein 3 (MIP3A) and interleukin-1 receptor 1 (IL1R1), but down-regulated fibroblast growth factor 13 (FGF13). The expression of FGF receptor activating protein 1 (FRAG1) and interleukin-3 receptor alpha (IL3RA) was down-regulated by GD3. Exposure to TNFα resulted in a dramatic up-regulation of IL3RA and chemokine ligand 2 (CCL2), both of which have been implicated in multiple sclerosis. Our results provide strong evidence that the expression of these genes might be critical links in the metabolic cascades leading to cell degeneration and death in the brain.
Collapse
|
8
|
Sluijs KFVD, Obregon C, Geiser TK, Mühlemann K, Nicod LP. Monocyte differentiation toward regulatory dendritic cells is not affected by respiratory syncytial virus-induced inflammatory mediators. Am J Respir Cell Mol Biol 2011; 44:655-64. [PMID: 20595462 DOI: 10.1165/rcmb.2010-0136oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Airway epithelial cells were shown to drive the differentiation of monocytes into dendritic cells (DCs) with a suppressive phenotype. In this study, we investigated the impact of virus-induced inflammatory mediator production on the development of DCs. Monocyte differentiation into functional DCs, as reflected by the expression of CD11c, CD123, BDCA-4, and DC-SIGN and the capacity to activate T cells, was similar for respiratory syncytial virus (RSV)-infected and mock-infected BEAS-2B and A549 cells. RSV-conditioned culture media resulted in a partially mature DC phenotype, but failed to up-regulate CD80, CD83, CD86, and CCR7, and failed to release proinflammatory mediators upon Toll-like receptor (TLR) triggering. Nevertheless, these DCs were able to maintain an antiviral response by the release of Type I IFN. Collectively, these data indicate that the airway epithelium maintains an important suppressive DC phenotype under the inflammatory conditions induced by infection with RSV.
Collapse
|
9
|
Exogenous control of the expression of Group I CD1 molecules competent for presentation of microbial nonpeptide antigens to human T lymphocytes. Clin Dev Immunol 2011; 2011:790460. [PMID: 21603161 PMCID: PMC3095450 DOI: 10.1155/2011/790460] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 01/12/2011] [Accepted: 01/19/2011] [Indexed: 12/21/2022]
Abstract
Group I CD1 (CD1a, CD1b, and CD1c) glycoproteins expressed on immature and mature dendritic cells present nonpeptide antigens (i.e., lipid or glycolipid molecules mainly of microbial origin) to T cells. Cytotoxic CD1-restricted T lymphocytes recognizing mycobacterial lipid antigens were found in tuberculosis patients. However, thanks to a complex interplay between mycobacteria and CD1 system, M. tuberculosis possesses a successful tactic based, at least in part, on CD1 downregulation to evade CD1-dependent immunity. On the ground of these findings, it is reasonable to hypothesize that modulation of CD1 protein expression by chemical, biological, or infectious agents could influence host's immune reactivity against M. tuberculosis-associated lipids, possibly affecting antitubercular resistance. This scenario prompted us to perform a detailed analysis of the literature concerning the effect of external agents on Group I CD1 expression in order to obtain valuable information on the possible strategies to be adopted for driving properly CD1-dependent immune functions in human pathology and in particular, in human tuberculosis.
Collapse
|
10
|
Immunoregulatory effects of the flavonol quercetin in vitro and in vivo. Eur J Nutr 2010; 50:163-72. [PMID: 20652710 DOI: 10.1007/s00394-010-0125-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 07/08/2010] [Indexed: 12/18/2022]
Abstract
PURPOSE Atherosclerosis is known to be an inflammatory disease. Dendritic cells (DCs) are essential for the regulation of the immune system. Up to 10% of the cells in atherosclerotic plaques are DCs. The cardiovascular protective effects of flavonoids (tea, wine) may be mediated by anti-inflammatory mechanisms that affect DC regulation. We aimed to characterize the impact of the flavonol quercetin on DC activity and differentiation in vitro and in vivo. METHODS For the in vitro experiments, we used murine DCs and endothelial cells to study adhesion properties. For all other experiments (DC phagocytosis capacity, DC maturation, DC differentiation (BDCA-1/-2) and NF-kB-activation), human monocyte-derived DCs were used. The cells were incubated with quercetin (10 μmol/L) ± oxLDL (10 μg/mL) between 24 and 48 h. For in vivo experiments, eight healthy male volunteers took 500 mg of quercetin twice daily over 4 weeks, five healthy male volunteers served as control. Before and after intake, blood samples were collected. Peripheral blood leukocytes were isolated (analyses of DC differentiation), and plasma was immediately frozen. RESULTS Quercetin reduced DC adhesion (-42%; p < 0.05) and expression of CD11a (-21%; p < 0.05). OxLDL-induced DC differentiation was partially inhibited by quercetin (BDCA-1-29%; BDCA-2-33%; p < 0.05). These effects were achieved by compensation of oxLDL-induced up-regulation of NF-kB by quercetin. The 4-week treatment with quercetin resulted in relevant plasma levels (2.47 μmol/L) and reduced BDCA-2 + DCs in the peripheral blood by 42% (p < 0.05) as well as systemic levels of the NO-synthase inhibitor asymmetric dimethylarginine (-31%, p < 0.05). CONCLUSION In vitro, quercetin reduced DC adhesion and oxLDL-induced DC differentiation. In vivo, quercetin reduced circulating plasmacytoid DCs and systemic ADMA-levels. The immunoregulatory effects of quercetin may contribute to the anti-atherosclerotic potential of flavonols.
Collapse
|
11
|
Bartosik-Psujek H, Tabarkiewicz J, Pocinska K, Radej S, Stelmasiak Z, Rolinski J. Immunomodulatory Effects of IFN-β and Lovastatin on Immunophenotype of Monocyte-Derived Dendritic Cells in Multiple Sclerosis. Arch Immunol Ther Exp (Warsz) 2010; 58:313-9. [DOI: 10.1007/s00005-010-0084-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 03/01/2010] [Indexed: 10/19/2022]
|
12
|
Yu CF, Peng WM, Oldenburg J, Hoch J, Bieber T, Limmer A, Hartmann G, Barchet W, Eis-Hübinger AM, Novak N. Human plasmacytoid dendritic cells support Th17 cell effector function in response to TLR7 ligation. THE JOURNAL OF IMMUNOLOGY 2009; 184:1159-67. [PMID: 20026744 DOI: 10.4049/jimmunol.0901706] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Signals involved in the commitment of Th17 differentiation are of substantial interest for our understanding of antimicrobial defense mechanisms and autoimmune disorders. Various ways in which myeloid dendritic cells modulate Th17 differentiation have been identified. However, although plasmacytoid dendritic cells (PDCs) are regarded as important players in antiviral/antimicrobial host defense and autoimmune diseases, a putative modulatory role of PDCs in Th17 differentiation has not yet been elucidated in detail. We demonstrated that PDCs are capable of promoting Th17 differentiation in response to TLR7 stimulation. Further, both the differentiation of Th17 cells from naive T cells and the amplification of Th17 effector functions of memory T cells are promoted by PDCs after TLR7 activation. Our data are of strong clinical relevance because TLR7 activation in PDCs might represent one of the missing links between innate and adaptive immune mechanisms and contribute to the amplification of Th17-driven autoimmune disorders as well as viral host defense.
Collapse
Affiliation(s)
- Chun-Feng Yu
- Department of Dermatology and Allergy, University Hospital, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Namdar A, Nikbin B, Ghabaee M, Bayati A, Izad M. Effect of IFN-beta therapy on the frequency and function of CD4(+)CD25(+) regulatory T cells and Foxp3 gene expression in relapsing-remitting multiple sclerosis (RRMS): a preliminary study. J Neuroimmunol 2009; 218:120-4. [PMID: 19932513 DOI: 10.1016/j.jneuroim.2009.10.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2009] [Revised: 09/29/2009] [Accepted: 10/21/2009] [Indexed: 11/29/2022]
Abstract
Interferon-beta (IFN-beta) is an immunomodulatory drug of choice to control relapsing-remitting multiple sclerosis (RR-MS), although its function is still unclear. A reduced suppressive function of CD4(+)CD25(+) regulatory T cells (T(reg)) has been shown in RR-MS patients. In this study, to understand the effect of IFN-ss on CD4(+)CD25(+) regulatory T cells, we analyzed the frequency and function of these cells and Foxp3 gene expression before and after treatment. We evaluated the frequency and function of CD4(+)CD25(+)Foxp3(+) regulatory T cells by flow cytometry and co-culture inhibition test respectively and gene expression of Foxp3 by real-time PCR in a longitudinal follow-up study in 18 relapsing-remitting MS patients. Our data revealed that IFN-beta significantly improved frequency and suppressive function of T(reg) cells (P<0.05) without any significant effect on gene expression of Foxp3 after 6 months. The results of the present study indicate that IFN-beta therapy in some of patients with RR-MS may restore function of regulatory T cells and control the unchecked immune cascade activity. Larger longitudinal studies on more MS patients are required to confirm our findings.
Collapse
Affiliation(s)
- Afshin Namdar
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | |
Collapse
|
14
|
Ross AC, Chen Q, Ma Y. Augmentation of antibody responses by retinoic acid and costimulatory molecules. Semin Immunol 2009; 21:42-50. [PMID: 18819820 PMCID: PMC2615053 DOI: 10.1016/j.smim.2008.08.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 08/13/2008] [Accepted: 08/18/2008] [Indexed: 11/22/2022]
Abstract
Antibody production is crucial for a successful vaccine response. Beyond the ability of vitamin A (VA) and its active metabolite, all-trans-retinoic acid (RA) to restore growth in VA-deficient animals, supplementation with VA and/or treatment with RA can augment antibody responses in both VA-deficient and VA-adequate animals. RA alone, and in combination with stimuli that are ligands for the Toll-like receptor family, can augment the adaptive immune response leading to a heightened primary antibody response, and a stronger recall response upon restimulation. Mechanisms may include regulation of cell populations, type 1/type 2 cytokines, and B cell-related transcription factors, leading to accelerated B cell maturation.
Collapse
Affiliation(s)
- A Catharine Ross
- Department of Nutritional Sciences, Pennsylvania State University, 110 Chandlee Laboratory, University Park, PA 16802, USA.
| | | | | |
Collapse
|
15
|
Martinez-Forero I, Pelaez A, Villoslada P. Pharmacogenomics of multiple sclerosis: in search for a personalized therapy. Expert Opin Pharmacother 2008; 9:3053-67. [DOI: 10.1517/14656560802515553] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
Sanna A, Huang YM, Arru G, Fois ML, Link H, Rosati G, Sotgiu S. Multiple sclerosis: reduced proportion of circulating plasmacytoid dendritic cells expressing BDCA-2 and BDCA-4 and reduced production of IL-6 and IL-10 in response to herpes simplex virus type 1. Mult Scler 2008; 14:1199-207. [PMID: 18653740 DOI: 10.1177/1352458508094401] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We hypothesized that autoaggressive immune responses observed in multiple sclerosis (MS) could be associated with an imbalance in proportion of immune cell subsets and in cytokine production in response to infection, including viruses. METHODS We collected blood mononuclear cells (MNC) from 23 patients with MS and 23 sex- and age-matched healthy controls (HC) from the island of Sardinia, Italy, where the prevalence of MS is extraordinarily high. Using flow cytometry, we studied MNC for expression of blood dendritic cell antigens (BDCA)-2 and BDCA-4 surface markers reflecting the proportion of plasmacytoid dendritic cells (pDC) that produce type I interferons (IFNs) after virus challenge and promote Th2/anti-inflammtory cytokine production. In parallel, pro-inflammatory (interleukin [IL]-2, IL-12, IFN-gamma), anti-inflammatory (IL-4, IL-10), and immuno-regulatory/pleiotropic cytokines (type I IFNs including IFN-alpha and beta, IL-6) were measured before and after an in vitro exposure to herpes simplex virus type 1 (HSV-1). RESULTS The subset of lineage negative (lin(-)), BDCA-2(+) cells was lower in patients with MS compared with HC (0.08 + or - 0.02% vs 0.24 + or - 0.02%; P < 0.001). A similar pattern was observed for lin(-)BDCA-4(+) cells (0.08 + or - 0.02% vs 0.17% + or - 0.03; P < 0.01). Spontaneous productions of IL-6 (45 + or - 10 pg/mL vs 140 + or - 26 pg/mL; P < 0.01) and IL-10 (17 + or - 0.4 pg/mL vs 21 + or - 1 pg/mL; P < 0.05) by MNC were lower in patients with MS compared with HC. Spontaneous production of IL-6 (6.5 + or - 0.15 pg/mL vs 21 + or - 5 pg/mL; P < 0.01 and IL-10 (11 + or - 1 pg/mL vs 14 + or - 3 pg/mL; P < 0.05) by pDC was also lower in patients with MS compared with HC. Exposure of MNC to HSV-1 showed, in both patients with MS and HC, increased production of IFN-alpha, IL-6, and IL-10 but decreased production of IL-4. In response to HSV-1 exposure, productions of IL-6 (165 +or - 28 pg/mL vs 325 + or - 35 pg/mL; P < 0.01) and IL-10 (27 +or - 3 vs 33 + or - 3 P < 0.05) by MNC as well as by pDC (IL-6: 28 + or - 7 vs 39 + or - 12 P < 0.05; IL-10: 14 + or - 1 vs 16 + or - 3 P < 0.05) were lower in patients with MS compared with HC. CONCLUSION The results implicate a new evidence for altered immune cells and reduced immune responses in response to viral challenge in MS.
Collapse
Affiliation(s)
- A Sanna
- Department of Neuroscience, Institute of Clinical Neurology, University of Sassari, Sassari, Italy.
| | | | | | | | | | | | | |
Collapse
|
17
|
de Andrés C, Aristimuño C, de Las Heras V, Martínez-Ginés ML, Bartolomé M, Arroyo R, Navarro J, Giménez-Roldán S, Fernández-Cruz E, Sánchez-Ramón S. Interferon beta-1a therapy enhances CD4+ regulatory T-cell function: an ex vivo and in vitro longitudinal study in relapsing-remitting multiple sclerosis. J Neuroimmunol 2006; 182:204-11. [PMID: 17157927 DOI: 10.1016/j.jneuroim.2006.09.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Revised: 09/19/2006] [Accepted: 09/26/2006] [Indexed: 10/23/2022]
Abstract
Interferon beta-1a (IFNâ-1a) has demonstrated efficacy in multiple sclerosis (MS), although its mechanism of action remains only partly understood. We evaluated the ex vivo and in vitro effects of IFNâ-1a (Rebif) on regulatory T-cell (T(Reg)) function in 22 relapsing-remitting MS patients and 16 healthy controls. T(Reg) function was significantly enhanced after 3 and 6 months of IFNbeta-1a therapy. Furthermore, there was a trend towards increasing proportions of total CD4(+)CD25(+) and CD4(+)CD25(+)GITR(+) T(Reg) after 6 months of IFNbeta-1a therapy when compared with baseline. In conclusion, IFNbeta-1a therapy enhances T(Reg) function, and this may be relevant in the inflammatory environment of MS lesions.
Collapse
Affiliation(s)
- Clara de Andrés
- Department of Neurology, University General Hospital Gregorio Marañón, Calle Doctor Esquerdo, 46, 28007 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
López C, Comabella M, Al-zayat H, Tintoré M, Montalban X. Altered maturation of circulating dendritic cells in primary progressive MS patients. J Neuroimmunol 2006; 175:183-91. [PMID: 16698091 DOI: 10.1016/j.jneuroim.2006.03.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Revised: 03/17/2006] [Accepted: 03/20/2006] [Indexed: 10/24/2022]
Abstract
We investigated the phenotype and frequency of circulating myeloid dendritic cells (MDC) and plasmacytoid DC (PDC) in 86 multiple sclerosis (MS) patients and 33 healthy controls (HC). The MS group comprised 20 patients with primary progressive MS (PPMS), 20 patients with secondary progressive MS (SPMS), and 46 patients with relapsing-remitting MS (RRMS) [23 treated with interferon-beta (IFN-beta)]. The frequency of circulating MDC and PDC, and the expression of CD83, CD123, CD80, CD86, and CD40 were analyzed by flow cytometry. The percentage of circulating MDC was decreased in patients with SPMS and PPMS. The expression of CD83, CD80, and CD86 was lower in PPMS patients. Treatment with IFN-beta induced the expression of CD123 in PDC and decreased the number of circulating MDC. These results suggest an impaired maturation state of DC in PPMS patients, and a beneficial effect of IFN-beta favouring the survival of PDC and promoting a Th2 environment.
Collapse
Affiliation(s)
- Cristina López
- Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d'Hebron, HUVH, Barcelona, Spain
| | | | | | | | | |
Collapse
|
19
|
Ma Y, Chen Q, Ross AC. Retinoic acid and polyriboinosinic:polyribocytidylic acid stimulate robust anti-tetanus antibody production while differentially regulating type 1/type 2 cytokines and lymphocyte populations. THE JOURNAL OF IMMUNOLOGY 2005; 174:7961-9. [PMID: 15944302 PMCID: PMC3843132 DOI: 10.4049/jimmunol.174.12.7961] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Retinoic acid (RA), a bioactive retinoid, and polyriboinosinic:polyribocytidylic acid (PIC) are known to promote immunity in vitamin A-deficient animals. In this study, we hypothesized that RA, PIC, and the combination can provide significant immunoadjuvant activity even in the vitamin A-adequate state. Six-week-old C57BL/6 mice were immunized with tetanus toxoid (TT) and treated with RA and/or PIC at priming in three independent studies of short and long duration. RA and PIC differentially regulated both primary and secondary anti-TT IgG isotypes, whereas the combination of RA + PIC stimulated the highest level of anti-TT IgG production and, concomitantly, a ratio of IgG1 to IgG2a similar to that of the control group. The regulation of Ab response was strongly associated with type 1/type 2 cytokine gene expression. Whereas RA reduced type 1 cytokines (IFN-gamma and IL-12), PIC enhanced both type 1 and type 2 cytokines (IL-4 and IL-12) and cytokine-related transcription factors. Despite the presence of PIC, the IL-4:IFN-gamma ratio was significantly elevated by RA. In addition, RA and/or PIC modulated NK/NKT cell populations and the level of expression of the costimulatory molecules CD80/CD86, evident 3 days after priming. Notably, the NKT:NK and CD80:CD86 ratios were correlated with the IL-4:IFN-gamma ratio, indicative of multiple converging modes of regulation. Overall, RA, PIC, and RA + PIC rapidly and differentially shaped the anti-tetanus Ig response. The robust, durable, and proportionate increase in all anti-TT IgG isotypes induced by RA + PIC suggests that this combination is promising as a means to enhance the Ab response to TT and similar vaccines.
Collapse
Affiliation(s)
- Yifan Ma
- Integrative Biosciences Nutritional Sciences Program, Pennsylvania State University, University Park, PA 16801
| | - Qiuyan Chen
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16801
| | - A. Catharine Ross
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16801
- Huck Institute for the Life Sciences, Pennsylvania State University, University Park, PA 16801
- Address correspondence and reprint requests to Dr. A. Catharine Ross, Department of Nutritional Sciences, Pennsylvania State University, 126-S Henderson Building, University Park, PA 16802.
| |
Collapse
|