1
|
Mann DL. The Emerging Field of Cardioimmunology: Past, Present and Foreseeable Future. Circ Res 2024; 134:1663-1680. [PMID: 38843286 PMCID: PMC11160976 DOI: 10.1161/circresaha.123.323656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/08/2024] [Indexed: 06/09/2024]
Abstract
Over the past 30 years, the field of cardioimmunology has moved from being dismissed as a field that was chasing an epiphenomenon of little biological consequence to a scientific discipline that is providing important new insights into the immunologic basis for hypertension, atherosclerosis, myocarditis, pericarditis, autoimmune heart disease, and heart failure. In this article, we will review the conceptual insights and technical breakthroughs that have allowed the field to move forward, as well as the clinical trials in the cardioimmunology space, to provide a historical context for the articles that will appear in the compendium that is focused on the interface between cardioimmunology, myocardial function, and disease.
Collapse
Affiliation(s)
- Douglas L Mann
- Cardiovascular Division, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
2
|
Zenewicz LA. IL-22 Binding Protein (IL-22BP) in the Regulation of IL-22 Biology. Front Immunol 2021; 12:766586. [PMID: 34868019 PMCID: PMC8634938 DOI: 10.3389/fimmu.2021.766586] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/25/2021] [Indexed: 01/21/2023] Open
Abstract
Cytokines are powerful mediators of inflammation. Consequently, their potency is regulated in many ways to protect the host. Several cytokines, including IL-22, have coordinating binding proteins or soluble receptors that bind to the cytokine, block the interaction with the cellular receptor, and thus prevent cellular signaling. IL-22 is a critical cytokine in the modulation of tissue responses during inflammation and is highly upregulated in many chronic inflammatory disease patients, including those with psoriasis, rheumatoid arthritis, and inflammatory bowel disease (IBD). In healthy individuals, low levels of IL-22 are secreted by immune cells, mainly in the gastrointestinal (GI) tract. However, much of this IL-22 is likely not biologically active due to the high levels of IL-22 binding protein (IL-22BP) produced by intestinal dendritic cells (DCs). IL-22BP is a soluble receptor homolog that binds to IL-22 with greater affinity than the membrane spanning receptor. Much is known regarding the regulation and function of IL-22 in health and disease. However, less is known about IL-22BP. In this review, we will focus on IL-22BP, including its regulation, role in IL-22 biology and inflammation, and promise as a therapeutic. IL-22 can be protective or pathogenic, depending on the context of inflammation. IL-22BP also has divergent roles. Ongoing and forthcoming studies will expand our knowledge of IL-22BP and IL-22 biology, and suggest that IL-22BP holds promise as a way to regulate IL-22 biology in patients with chronic inflammatory disease.
Collapse
Affiliation(s)
- Lauren A. Zenewicz
- Department of Microbiology and Immunology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
3
|
Barrett D, Bilic S, Chyung Y, Cote SM, Iarrobino R, Kacena K, Kalra A, Long K, Nomikos G, Place A, Still JG, Vrishabhendra L. A Randomized Phase 1 Safety, Pharmacokinetic and Pharmacodynamic Study of the Novel Myostatin Inhibitor Apitegromab (SRK-015): A Potential Treatment for Spinal Muscular Atrophy. Adv Ther 2021; 38:3203-3222. [PMID: 33963971 PMCID: PMC8189951 DOI: 10.1007/s12325-021-01757-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/20/2021] [Indexed: 12/31/2022]
Abstract
Introduction Apitegromab (SRK-015) is an anti-promyostatin monoclonal antibody under development to improve motor function in patients with spinal muscular atrophy, a rare neuromuscular disease. This phase 1 double-blind, placebo-controlled study assessed safety, pharmacokinetic parameters, pharmacodynamics (serum latent myostatin), and immunogenicity of single and multiple ascending doses of apitegromab in healthy adult subjects. Methods Subjects were administered single intravenous ascending doses of apitegromab of 1, 3, 10, 20, 30 mg/kg or placebo, and multiple intravenous ascending doses of apitegromab of 10, 20, 30 mg/kg or placebo. Results Following single ascending doses, the pharmacokinetic parameters of apitegromab appeared to be similar across all dose groups, following a biphasic pattern of decline in the concentration–time curve. The mean apparent terminal t1/2 after single intravenous doses of apitegromab ranged from 24 to 31 days across dose groups. Dose-related increases were observed in Cmax following multiple ascending doses. Single and multiple apitegromab doses resulted in dose-dependent and sustained increases in serum latent myostatin, indicating robust target engagement. Apitegromab was safe and well tolerated, on the basis of the adverse event (AE) profile with no clinically meaningful changes in baseline vital signs, electrocardiograms, or clinical laboratory parameters and no anti-drug antibody formation. Conclusion These results support continued investigation of apitegromab for the treatment of patients with milder forms (type 2 and 3) of spinal muscular atrophy.
Collapse
|
4
|
Sahin D, Arenas-Ramirez N, Rath M, Karakus U, Hümbelin M, van Gogh M, Borsig L, Boyman O. An IL-2-grafted antibody immunotherapy with potent efficacy against metastatic cancer. Nat Commun 2020; 11:6440. [PMID: 33353953 PMCID: PMC7755894 DOI: 10.1038/s41467-020-20220-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/13/2020] [Indexed: 11/27/2022] Open
Abstract
Modified interleukin-2 (IL-2) formulations are being tested in cancer patients. However, IL-2 immunotherapy damages IL-2 receptor (IL-2R)-positive endothelial cells and stimulates IL-2Rα (CD25)-expressing lymphocytes that curtail anti-tumor responses. A first generation of IL-2Rβ (CD122)-biased IL-2s addressed some of these drawbacks. Here, we present a second-generation CD122-biased IL-2, developed by splitting and permanently grafting unmutated human IL-2 (hIL-2) to its antigen-binding groove on the anti-hIL-2 monoclonal antibody NARA1, thereby generating NARA1leukin. In comparison to hIL-2/NARA1 complexes, NARA1leukin shows a longer in vivo half-life, completely avoids association with CD25, and more potently stimulates CD8+ T and natural killer cells. These effects result in strong anti-tumor responses in various pre-clinical cancer models, whereby NARA1leukin consistently surpasses the efficacy of hIL-2/NARA1 complexes in controlling metastatic disease. Collectively, NARA1leukin is a CD122-biased single-molecule construct based on unmutated hIL-2 with potent efficacy against advanced malignancies. IL-2/anti-IL-2 complexes have been proposed to curtail the severe adverse effects associated with IL-2 immunotherapy. Here, the authors, by integrating unmutated human IL-2 in the antigen binding groove of an anti-IL-2 monoclonal antibody, generate a CD122-biased fusion protein that prevents binding of IL-2 to CD25 and promotes anti-tumor immune response in several preclinical metastatic cancer models.
Collapse
Affiliation(s)
- Dilara Sahin
- Department of Immunology, University Hospital Zurich, CH-8091, Zurich, Switzerland
| | | | - Matthias Rath
- Department of Immunology, University Hospital Zurich, CH-8091, Zurich, Switzerland
| | - Ufuk Karakus
- Department of Immunology, University Hospital Zurich, CH-8091, Zurich, Switzerland
| | - Monika Hümbelin
- Department of Immunology, University Hospital Zurich, CH-8091, Zurich, Switzerland
| | - Merel van Gogh
- Institute of Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Lubor Borsig
- Institute of Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, CH-8091, Zurich, Switzerland. .,Faculty of Medicine, University of Zurich, CH-8006, Zurich, Switzerland.
| |
Collapse
|
5
|
Abstract
Heart failure exhibits remarkable pathophysiologic heterogeneity. A large body of evidence suggests that regardless of the underlying etiology, heart failure is associated with induction of cytokines and chemokines that may contribute to the pathogenesis of adverse remodeling, and systolic and diastolic dysfunction. The pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-6 have been extensively implicated in the pathogenesis of heart failure. Inflammatory cytokines modulate phenotype and function of all myocardial cells, suppressing contractile function in cardiomyocytes, inducing inflammatory activation in macrophages, stimulating microvascular inflammation and dysfunction, and promoting a matrix-degrading phenotype in fibroblasts. Moreover, cytokine-induced growth factor synthesis may exert chronic fibrogenic actions contributing to the pathogenesis of heart failure with preserved ejection fraction (HFpEF). In addition to their role in adverse cardiac remodeling, some inflammatory cytokines may also exert protective actions on cardiomyocytes under conditions of stress. Chemokines, such as CCL2, are also upregulated in failing hearts and may stimulate recruitment of pro-inflammatory leukocytes, promoting myocardial injury, fibrotic remodeling, and dysfunction. Although experimental evidence suggests that cytokine and chemokine targeting may hold therapeutic promise in heart failure, clinical translation remains challenging. This review manuscript summarizes our knowledge on the role of TNF-α, IL-1, IL-6, and CCL2 in the pathogenesis of heart failure, and discusses the promises and challenges of targeted anti-cytokine therapy. Dissection of protective and maladaptive cellular actions of cytokines in the failing heart, and identification of patient subsets with overactive or dysregulated myocardial inflammatory responses are required for design of successful therapeutic approaches.
Collapse
|
6
|
Van Linthout S, Tschöpe C. The Quest for Antiinflammatory and Immunomodulatory Strategies in Heart Failure. Clin Pharmacol Ther 2019; 106:1198-1208. [PMID: 31544235 DOI: 10.1002/cpt.1637] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022]
Abstract
Intensive research over the last 3 decades has unequivocally demonstrated the relevance of inflammation in heart failure (HF). Despite our current and ever increasing knowledge about inflammation, the clinical success of antiinflammatory and immunomodulatory therapies in HF is still limited. This review outlines the complexity and diversity of inflammation, its reciprocal interaction with HF, and addresses future perspectives, calling for immunomodulatory therapies that are specific for factors that activate the immune system without the risk of nonspecific immune suppression.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Campus Virchow Clinic, Charité University Medicine Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Campus Virchow Clinic, Charité University Medicine Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site, Berlin, Germany.,Department of Cardiology, Campus Virchow Klinikum, Charité, University Medicine Berlin, Berlin, Germany
| |
Collapse
|
7
|
Filardi T, Ghinassi B, Di Baldassarre A, Tanzilli G, Morano S, Lenzi A, Basili S, Crescioli C. Cardiomyopathy Associated with Diabetes: The Central Role of the Cardiomyocyte. Int J Mol Sci 2019; 20:ijms20133299. [PMID: 31284374 PMCID: PMC6651183 DOI: 10.3390/ijms20133299] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022] Open
Abstract
The term diabetic cardiomyopathy (DCM) labels an abnormal cardiac structure and performance due to intrinsic heart muscle malfunction, independently of other vascular co-morbidity. DCM, accounting for 50%–80% of deaths in diabetic patients, represents a worldwide problem for human health and related economics. Optimal glycemic control is not sufficient to prevent DCM, which derives from heart remodeling and geometrical changes, with both consequences of critical events initially occurring at the cardiomyocyte level. Cardiac cells, under hyperglycemia, very early undergo metabolic abnormalities and contribute to T helper (Th)-driven inflammatory perturbation, behaving as immunoactive units capable of releasing critical biomediators, such as cytokines and chemokines. This paper aims to focus onto the role of cardiomyocytes, no longer considered as “passive” targets but as “active” units participating in the inflammatory dialogue between local and systemic counterparts underlying DCM development and maintenance. Some of the main biomolecular/metabolic/inflammatory processes triggered within cardiac cells by high glucose are overviewed; particular attention is addressed to early inflammatory cytokines and chemokines, representing potential therapeutic targets for a prompt early intervention when no signs or symptoms of DCM are manifesting yet. DCM clinical management still represents a challenge and further translational investigations, including studies at female/male cell level, are warranted.
Collapse
Affiliation(s)
- Tiziana Filardi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti and Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Susanna Morano
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, "Sapienza" University, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Clara Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Sciences, University of Rome "Foro Italico", Piazza L. de Bosis 6, 00135 Rome, Italy.
| |
Collapse
|
8
|
Long KK, O’Shea KM, Khairallah RJ, Howell K, Paushkin S, Chen KS, Cote SM, Webster MT, Stains JP, Treece E, Buckler A, Donovan A. Specific inhibition of myostatin activation is beneficial in mouse models of SMA therapy. Hum Mol Genet 2019; 28:1076-1089. [PMID: 30481286 PMCID: PMC6423420 DOI: 10.1093/hmg/ddy382] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 12/22/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by loss of α-motor neurons, leading to profound skeletal muscle atrophy. Patients also suffer from decreased bone mineral density and increased fracture risk. The majority of treatments for SMA, approved or in clinic trials, focus on addressing the underlying cause of disease, insufficient production of full-length SMN protein. While restoration of SMN has resulted in improvements in functional measures, significant deficits remain in both mice and SMA patients following treatment. Motor function in SMA patients may be additionally improved by targeting skeletal muscle to reduce atrophy and improve muscle strength. Inhibition of myostatin, a negative regulator of muscle mass, offers a promising approach to increase muscle function in SMA patients. Here we demonstrate that muSRK-015P, a monoclonal antibody which specifically inhibits myostatin activation, effectively increases muscle mass and function in two variants of the pharmacological mouse model of SMA in which pharmacologic restoration of SMN has taken place either 1 or 24 days after birth to reflect early or later therapeutic intervention. Additionally, muSRK-015P treatment improves the cortical and trabecular bone phenotypes in these mice. These data indicate that preventing myostatin activation has therapeutic potential in addressing muscle and bone deficiencies in SMA patients. An optimized variant of SRK-015P, SRK-015, is currently in clinical development for treatment of SMA.
Collapse
Affiliation(s)
| | | | | | - Kelly Howell
- SMA Foundation, 888 7th Avenue #400, New York, NY
| | | | - Karen S Chen
- SMA Foundation, 888 7th Avenue #400, New York, NY
| | - Shaun M Cote
- Scholar Rock Inc., 620 Memorial Drive, Cambridge, MA
| | | | - Joseph P Stains
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erin Treece
- Scholar Rock Inc., 620 Memorial Drive, Cambridge, MA
| | - Alan Buckler
- Scholar Rock Inc., 620 Memorial Drive, Cambridge, MA
| | | |
Collapse
|
9
|
Spohn G, Arenas-Ramirez N, Bouchaud G, Boyman O. Endogenous polyclonal anti–IL-1 antibody responses potentiate IL-1 activity during pathogenic inflammation. J Allergy Clin Immunol 2017; 139:1957-1965.e3. [DOI: 10.1016/j.jaci.2016.09.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 08/27/2016] [Accepted: 09/06/2016] [Indexed: 10/20/2022]
|
10
|
Abstract
Elevated levels of inflammatory mediators have been identified in patients with heart failure, including heart failure with reduced and preserved ejection fraction, as well as acute decompensated heart failure. Moreover, experimental studies have shown repeatedly that activation of inflammation in the heart provokes left ventricular remodeling and left ventricular dysfunction. Nonetheless, phase III clinical trials that have attempted to antagonize inflammatory mediators have been negative with respect to the primary end points of the trials, and in some patients, resulted in worsening heart failure or death. The following review will discuss how recent developments in the field of innate immunity have advanced our understanding of the role of inflammation in the pathogenesis of heart failure and will discuss the negative outcomes of the existing clinical trials in light of this new information.
Collapse
Affiliation(s)
- Douglas L Mann
- From the Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
11
|
Bhan U, Podsiad AB, Kovach MA, Ballinger MN, Keshamouni V, Standiford TJ. Linezolid has unique immunomodulatory effects in post-influenza community acquired MRSA pneumonia. PLoS One 2015; 10:e0114574. [PMID: 25635685 PMCID: PMC4312022 DOI: 10.1371/journal.pone.0114574] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 11/11/2014] [Indexed: 12/17/2022] Open
Abstract
Introduction Post influenza pneumonia is a leading cause of mortality and morbidity, with mortality rates approaching 60% when bacterial infections are secondary to multi-drug resistant (MDR) pathogens. Staphylococcus aureus, in particular community acquired MRSA (cMRSA), has emerged as a leading cause of post influenza pneumonia. Hypothesis Linezolid (LZD) prevents acute lung injury in murine model of post influenza bacterial pneumonia Methods Mice were infected with HINI strain of influenza and then challenged with cMRSA at day 7, treated with antibiotics (LZD or Vanco) or vehicle 6 hours post bacterial challenge and lungs and bronchoalveolar lavage fluid (BAL) harvested at 24 hours for bacterial clearance, inflammatory cell influx, cytokine/chemokine analysis and assessment of lung injury. Results Mice treated with LZD or Vanco had lower bacterial burden in the lung and no systemic dissemination, as compared to the control (no antibiotic) group at 24 hours post bacterial challenge. As compared to animals receiving Vanco, LZD group had significantly lower numbers of neutrophils in the BAL (9×103 vs. 2.3×104, p < 0.01), which was associated with reduced levels of chemotactic chemokines and inflammatory cytokines KC, MIP-2, IFN-γ, TNF-α and IL-1β in the BAL. Interestingly, LZD treatment also protected mice from lung injury, as assessed by albumin concentration in the BAL post treatment with H1N1 and cMRSA when compared to vanco treatment. Moreover, treatment with LZD was associated with significantly lower levels of PVL toxin in lungs. Conclusion Linezolid has unique immunomodulatory effects on host inflammatory response and lung injury in a murine model of post-viral cMRSA pneumonia.
Collapse
Affiliation(s)
- Urvashi Bhan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Amy B. Podsiad
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Melissa A. Kovach
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Megan N. Ballinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Ohio State University, Columbus, Ohio, United States of America
| | - Venkateshwar Keshamouni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| |
Collapse
|
12
|
Isik G, van Montfort T, Chung NPY, Moore JP, Sanders RW. Autoantibodies induced by chimeric cytokine-HIV envelope glycoprotein immunogens. THE JOURNAL OF IMMUNOLOGY 2014; 192:4628-35. [PMID: 24729614 DOI: 10.4049/jimmunol.1303401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cytokines are often used as adjuvants to increase the immunogenicity of vaccines because they can improve the immune response and/or direct it into a desired direction. As an alternative to codelivering Ags and cytokines separately, they can be fused into a composite protein, with the advantage that both moieties act on the same immune cells. The HIV-1 envelope glycoprotein (Env) spike, located on the outside of virus particles and the only relevant protein for the induction of neutralizing Abs, is poorly immunogenic. The induction of anti-Env Abs can be improved by coupling Env proteins to costimulatory molecules such as a proliferation inducing ligand (APRIL). In this study, we evaluated the immunogenicity of chimeric molecules containing uncleaved Env gp140 fused to the species-matched cytokines IL-21 or GM-CSF in rabbits and mice. Each cytokine was either fused to the C terminus of Env or embedded within Env at the position of the variable loops 1 and 2. The cytokine components of the chimeric Env-GM-CSF and Env-IL-21 molecules were functional in vitro, but none of the Env-cytokine fusion proteins resulted in improved Ab responses in vivo. Both the Env-GM-CSF and the Env-IL-21 molecules induced strong anticytokine Ab responses in both test species. These autoimmune responses were independent of the location of the cytokine in the chimeric Env molecules in that they were induced by cytokines inserted within the variable loops 1 and 2 of Env or fused to its C terminus. The induction of undesired autoimmune responses should be considered when using cytokines as costimulatory molecules in fusion proteins.
Collapse
Affiliation(s)
- Gözde Isik
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, 1100 DE Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
13
|
Fritz-French C, Shawahna R, Ward JE, Maroun LE, Tyor WR. The recombinant vaccinia virus gene product, B18R, neutralizes interferon alpha and alleviates histopathological complications in an HIV encephalitis mouse model. J Interferon Cytokine Res 2014; 34:510-7. [PMID: 24564363 DOI: 10.1089/jir.2013.0072] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Interferon-alpha (IFN-α) has been identified as a neurotoxin that plays a prominent role in human immunodeficiency virus (HIV)-associated neurocognitive disorders and HIV encephalitis (HIVE) pathology. IFN-α is associated with cognitive dysfunction in other inflammatory diseases where IFN-α is upregulated. Trials of monoclonal anti-IFN-α antibodies have been generally disappointing possibly due to high specificity to limited IFN-α subtypes and low affinity. We investigated a novel IFN-α inhibitor, B18R, in an HIVE/severe combined immunodeficiency (SCID) mouse model. Immunostaining for B18R in systemically treated HIVE/SCID mice suggested the ability of B18R to cross the blood-brain barrier (BBB). Real-time PCR indicated that B18R treatment resulted in a decrease in gene expression associated with IFN-α signaling in the brain. Mice treated with B18R were found to have decreased mouse mononuclear phagocytes and significant retention of neuronal arborization compared to untreated HIVE/SCID mice. Increased mononuclear phagocytes and decreased neuronal arborization are key features of HIVE. These results suggest that B18R crosses the BBB, blocks IFN-α signaling, and it prevents key features of HIVE pathology. These data suggest that the high affinity and broad IFN-α subtype specificity of B18R make it a viable alternative to monoclonal antibodies for the inhibition of IFN-α in the immune-suppressed environment.
Collapse
Affiliation(s)
- Cari Fritz-French
- 1 Department of Neurology, Emory University School of Medicine , Atlanta, Georgia
| | | | | | | | | |
Collapse
|
14
|
|
15
|
Abstract
Cytokines and their receptors are involved in the pathophysiology of many diseases. Here we present a detailed review on cytokines, receptors and signalling routes, and show that one important lesson from cytokine biology is the complex and diverse regulation of cytokine activity. The activity of cytokines is controlled at the level of transcription, translation, storage, processing, posttranslational modification, trapping, binding by soluble proteins, and receptor number and/or function. Translation of this diverse regulation in strategies aimed at the control of cytokine activity will result in the development of more specific and selective drugs to treat diseases.
Collapse
|
16
|
Wrigley BJ, Lip GYH, Shantsila E. The role of monocytes and inflammation in the pathophysiology of heart failure. Eur J Heart Fail 2011; 13:1161-71. [PMID: 21952932 DOI: 10.1093/eurjhf/hfr122] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
There is growing evidence to support an important role of inflammation in the underlying pathophysiology of heart failure (HF). Indeed, inflammatory cytokine levels are well recognized to be increased in patients with left ventricular dysfunction and appear to have prognostic implications. Monocytes play a pivotal role in the inflammatory cascade and are a major source of both pro- and anti-inflammatory cytokines. They are intimately involved in tissue damage and repair and an imbalance of these processes may have detrimental consequences for the failing myocardium. Importantly, monocytes comprise of distinct subsets with different cell surface markers and functional characteristics and this heterogeneity may be important in understanding their specific role in HF. In HF, monocyte activation involves interplay between pattern recognition molecules, endotoxins, cytokines, and acute phase proteins. Activated monocytes migrate to the myocardium in response to powerful chemokines, where they must then attach to the endothelial wall before infiltrating into the myocardium itself. This review article aims to discuss the role of monocytes and inflammation in HF, focusing on monocyte activation, mobilisation, recruitment and endothelial adherence, as well as the effects they may have on myocardial performance. The therapeutic modulation of inflammation and monocyte activation in HF treatment will also be reviewed.
Collapse
Affiliation(s)
- Benjamin J Wrigley
- University of Birmingham Centre for Cardiovascular Sciences, City Hospital, Birmingham B18 7QH, UK
| | | | | |
Collapse
|
17
|
Rattigan Y, Hsu JM, Mishra PJ, Glod J, Banerjee D. Interleukin 6 mediated recruitment of mesenchymal stem cells to the hypoxic tumor milieu. Exp Cell Res 2010; 316:3417-24. [PMID: 20633553 DOI: 10.1016/j.yexcr.2010.07.002] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 06/30/2010] [Accepted: 07/03/2010] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem cells (MSCs) are a heterogeneous population of non-hematopoietic precursor cells predominantly found in the bone marrow. They have been recently reported to home towards the hypoxic tumor microenvironment in vivo. Interleukin-6 is a multifunctional cytokine normally involved in the regulation of the immune and inflammatory response. In addition to its normal function, IL-6 signaling has been implicated in tumorigenesis. Solid tumors develop hypoxia as a result of inadequate O(2) supply. Interestingly, tumor types with increased levels of hypoxia are known to have increased resistance to chemotherapy as well as increased metastatic potential. Here, we present evidence that under hypoxic conditions (1.5% O(2)) breast cancer cells secrete high levels of IL-6, which serve to activate and attract MSCs. We now report that secreted IL-6 acts in a paracrine fashion on MSCs stimulating the activation of both Stat3 and MAPK signaling pathways to enhance migratory potential and cell survival. Inhibition of IL-6 signaling utilizing neutralizing antibodies leads to attenuation of MSC migration. Specifically, increased migration is dependent on IL-6 signaling through the IL-6 receptor. Collectively, our data demonstrate that hypoxic tumor cells specifically recruit MSCs, which through activation of signaling and survival pathways facilitate tumor progression.
Collapse
Affiliation(s)
- Yanique Rattigan
- Department of Pharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ 08903, USA
| | | | | | | | | |
Collapse
|
18
|
Interleukin-1 Trap Attenuates Cardiac Remodeling After Experimental Acute Myocardial Infarction in Mice. J Cardiovasc Pharmacol 2010; 55:117-22. [DOI: 10.1097/fjc.0b013e3181c87e53] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
IL-2/anti-IL-2 antibody complexes show strong biological activity by avoiding interaction with IL-2 receptor alpha subunit CD25. Proc Natl Acad Sci U S A 2010; 107:2171-6. [PMID: 20133862 DOI: 10.1073/pnas.0909384107] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
IL-2 is crucial to T cell homeostasis, especially of CD4(+) T regulatory cells and memory CD8(+) cells, as evidenced by vigorous proliferation of these cells in vivo following injections of superagonist IL-2/anti-IL-2 antibody complexes. The mechanism of IL-2/anti-IL-2 antibody complexes is unknown owing to a lack of understanding of IL-2 homeostasis. We show that IL-2 receptor alpha (CD25) plays a crucial role in IL-2 homeostasis. Thus, prolongation of IL-2 half-life and blocking of CD25 using antibodies or CD25-deficient mice led in combination, but not alone, to vigorous IL-2-mediated T cell proliferation, similar to IL-2/anti-IL-2 antibody complexes. These data suggest an unpredicted role for CD25 in IL-2 homeostasis.
Collapse
|
20
|
Tomala J, Chmelova H, Mrkvan T, Rihova B, Kovar M. In Vivo Expansion of Activated Naive CD8+ T Cells and NK Cells Driven by Complexes of IL-2 and Anti-IL-2 Monoclonal Antibody As Novel Approach of Cancer Immunotherapy. THE JOURNAL OF IMMUNOLOGY 2009; 183:4904-12. [DOI: 10.4049/jimmunol.0900284] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
21
|
Abstract
Immunotherapy in the critically ill is an appealing notion because of the apparent abnormal immune and inflammatory responses seen in so many patients. The administration of a medication that could alter immune responses and decrease mortality in patients with sepsis could represent a ‘magic bullet’. Various approaches have been tried over the last 20 yr: steroids; anti-endotoxin or anti-cytokine antibodies; cytokine receptor antagonists; and other agents with immune-modulating side-effects. However, in some respects, research along these lines has been unsuccessful or disappointing at best. The current state of knowledge is summarized with particular reference to sepsis and the acute respiratory distress syndrome.
Collapse
Affiliation(s)
- N R Webster
- Anaesthesia and Intensive Care, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | | |
Collapse
|
22
|
von Haehling S, Lainscak M, Springer J, Anker SD. Cardiac cachexia: a systematic overview. Pharmacol Ther 2008; 121:227-52. [PMID: 19061914 DOI: 10.1016/j.pharmthera.2008.09.009] [Citation(s) in RCA: 258] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2008] [Accepted: 09/03/2008] [Indexed: 01/10/2023]
Abstract
Cardiac cachexia as a terminal stage of chronic heart failure carries a poor prognosis. The definition of this clinical syndrome has been a matter of debate in recent years. This review describes the ongoing discussion about this issue and the complex pathophysiology of cardiac cachexia and chronic heart failure with particular focus on immunological, metabolic, and hormonal aspects at the intracellular and extracellular level. These include regulators such as neuropeptide Y, leptin, melanocortins, ghrelin, growth hormone, and insulin. The regulation of feeding is discussed as are nutritional aspects in the treatment of the disease. The mechanisms of wasting in different body compartments are described. Moreover, we discuss several therapeutic approaches. These include appetite stimulants like megestrol acetate, medroxyprogesterone acetate, and cannabinoids. Other drug classes of interest comprise angiotensin-converting enzyme inhibitors, beta-blockers, anabolic steroids, beta-adrenergic agonists, anti-inflammatory substances, statins, thalidomide, proteasome inhibitors, and pentoxifylline.
Collapse
Affiliation(s)
- Stephan von Haehling
- Applied Cachexia Research, Department of Cardiology, Charité Medical School, Campus Virchow-Klinikum, Berlin, Germany.
| | | | | | | |
Collapse
|
23
|
Boyman O, Ramsey C, Kim DM, Sprent J, Surh CD. IL-7/Anti-IL-7 mAb Complexes Restore T Cell Development and Induce Homeostatic T Cell Expansion without Lymphopenia. THE JOURNAL OF IMMUNOLOGY 2008; 180:7265-75. [DOI: 10.4049/jimmunol.180.11.7265] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
24
|
High titer autoantibodies to GM-CSF in patients with AML, CML and MDS are associated with active disease. Leukemia 2008; 22:783-90. [PMID: 18216869 DOI: 10.1038/sj.leu.2405104] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Antibodies to granulocyte-macrophage colony-stimulating factor (GM-CSF) can be induced when GM-CSF is used as an adjuvant to solid tumor vaccination. Neutralizing anti-GM-CSF IgG has been associated with pulmonary alveolar proteinosis (PAP), and secondary PAP has been linked to myeloid leukemia. We studied 69 patients with acute myeloid leukemia, chronic myeloid leukemia and myelodysplastic syndrome, including 19 patients who received GM-CSF with peptide antigen and incomplete Freund's adjuvant in a vaccine trial for the presence or induction of anti-GM-CSF antibodies. Anti-GM-CSF IgG were present in 36 (52%) patients with myeloid leukemia compared to only 1 of 33 (3%) healthy subjects (P=0.008) and in none of 6 patients with lymphoid leukemia (P=0.0001). Antibody titers were unaffected by vaccination. Anti-GM-CSF IgA and IgM were found in 33 and 20% of patients, respectively; IgA from two patients neutralized GM-CSF. Strikingly, while anti-GM-CSF IgG titers were higher in patients with active disease (n=52) versus those in complete remission (n=14, P=0.0009), GM-CSF expression was not increased in either group. These data are first to show that anti-GM-CSF antibodies of multiple isotypes are present in patients with active myeloid leukemia without PAP and may be useful markers of disease activity.
Collapse
|
25
|
Buelow R, van Schooten W. The future of antibody therapy. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2007:83-106. [PMID: 17824183 DOI: 10.1007/2789_2007_040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Antibodies have been used successfully as therapeutics for over 100 years. The successful development of therapeutic human(ized) monoclonal antibodies (mAbs) in the last 20 years has demonstrated the potency of mAbs but also revealed some of their limitations. Studies in animals and humans demonstrated that it is possible to overcome some of these limitations using mixtures of mAbs or polyclonal antibody (pAb) preparations. pAbs from human and animal plasma are efficacious and safe therapeutics for the treatment of many diseases. Novel technologies are being developed for the production of human pAbs in genetically engineered animals. Immunization of such animals should allow the production of effective and safe high-titer antibody preparations for the treatment of infectious diseases, cancer, and autoimmunity.
Collapse
Affiliation(s)
- R Buelow
- Therapeutic Human Polyclonals Inc., 2105 Landings Drive, 94043, Mountain View, USA.
| | | |
Collapse
|
26
|
Hedrich O, Finley J, Konstam MA, Udelson JE. Novel neurohormonal antagonist strategies: vasopressin antagonism, anticytokine therapy, and endothelin antagonism in patients who have heart failure. Heart Fail Clin 2007; 1:103-27. [PMID: 17386838 DOI: 10.1016/j.hfc.2004.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Olaf Hedrich
- Tufts-New England Medical Center and Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | |
Collapse
|
27
|
Boyman O, Surh CD, Sprent J. Potential use of IL-2/anti-IL-2 antibody immune complexes for the treatment of cancer and autoimmune disease. Expert Opin Biol Ther 2007; 6:1323-31. [PMID: 17223740 DOI: 10.1517/14712598.6.12.1323] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Initially discovered as a potent T cell proliferation factor, IL-2 was soon used for cancer immunotherapy, especially for metastatic melanoma and renal cell carcinoma; however, the severe side effects of IL-2 therapy, plus the negative role of IL-2 in maintaining of CD4+ CD25+ T regulatory cells (Tregs), has somewhat dampened enthusiasm for using IL-2 in immunotherapy. This opinion article discusses the possibility of combining IL-2 with certain anti-IL-2 antibodies for reducing the dose of IL-2 needed and preferentially stimulating effector T cells, but not Tregs, an approach that might provide an improved strategy for anticancer immunotherapy. Alternatively, complexes of IL-2 with other anti-IL-2 antibodies can selectively stimulate Tregs and could, therefore, be useful for treating autoimmune diseases.
Collapse
Affiliation(s)
- Onur Boyman
- University Hospital of Lausanne (CHUV), Division of Immunology and Allergy, BT06 609, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland.
| | | | | |
Collapse
|
28
|
Garcia-Roca P, Mancilla-Ramirez J, Santos-Segura A, Fernández-Avilés M, Calderon-Jaimes E. Linezolid diminishes inflammatory cytokine production from human peripheral blood mononuclear cells. Arch Med Res 2006; 37:31-5. [PMID: 16314183 DOI: 10.1016/j.arcmed.2005.05.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Accepted: 05/26/2005] [Indexed: 11/22/2022]
Abstract
BACKGROUND Active peptides produced by monocytes, in response to endotoxin, initiate and maintain the acute phase of inflammatory response. Some antibiotics have been reported to have immunomodulatory effects in addition to their antimicrobial activity. We examined the effect of linezolid on cytokine synthesis. METHODS The modulatory effects of erythromycin and linezolid were evaluated in LPS-stimulated human peripheral blood mononuclear cells (PBMCs). Blood was obtained by venipuncture from healthy donor volunteers. PBMCs were separated by Ficoll-Paque. More than 90% of the cells were monocytes as determined by esterase staining. Cells were incubated in the presence of LPS, with or without various concentrations of erythromycin and linezolid. The concentration of each cytokine was determined by ELISA with commercially available reagents. RESULTS The two drugs suppressed significantly the synthesis of the cytokines tested in a concentration-dependent manner. CONCLUSIONS These data indicate that antibacterial agents may modify acute phase inflammatory response through their effects on cytokines synthesis by monocytes.
Collapse
Affiliation(s)
- Pilar Garcia-Roca
- Departamento de Inmunoquímica y Biología Celular, Hospital Infantil de México, Federico Gómez, México, D.F., México
| | | | | | | | | |
Collapse
|
29
|
Boyman O, Kovar M, Rubinstein MP, Surh CD, Sprent J. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science 2006; 311:1924-7. [PMID: 16484453 DOI: 10.1126/science.1122927] [Citation(s) in RCA: 707] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Interleukin-2 (IL-2), which is a growth factor for T lymphocytes, can also sometimes be inhibitory. Thus, the proliferation of CD8+ T cells in vivo is increased after the injection of a monoclonal antibody that is specific for IL-2 (IL-2 mAb), perhaps reflecting the removal of IL-2-dependent CD4+ T regulatory cells (T regs). Instead, we show here that IL-2 mAb augments the proliferation of CD8+ cells in mice simply by increasing the biological activity of preexisting IL-2 through the formation of immune complexes. When coupled with recombinant IL-2, some IL-2/IL-2 mAb complexes cause massive (>100-fold) expansion of CD8+ cells in vivo, whereas others selectively stimulate CD4+ T regs. Thus, different cytokine-antibody complexes can be used to selectively boost or inhibit the immune response.
Collapse
Affiliation(s)
- Onur Boyman
- Department of Immunology, IMM4, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
30
|
Rossi JF, Fegueux N, Lu ZY, Legouffe E, Exbrayat C, Bozonnat MC, Navarro R, Lopez E, Quittet P, Daures JP, Rouillé V, Kanouni T, Widjenes J, Klein B. Optimizing the use of anti-interleukin-6 monoclonal antibody with dexamethasone and 140 mg/m2 of melphalan in multiple myeloma: results of a pilot study including biological aspects. Bone Marrow Transplant 2006; 36:771-9. [PMID: 16113665 PMCID: PMC2034604 DOI: 10.1038/sj.bmt.1705138] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Interleukin-6 (IL-6) is a major survival factor for multiple myeloma (MM) cells preventing apoptosis induced by dexamethasone (DEX) or chemotherapy. In all, 24 consecutive patients with MM in first-line therapy received DEX for 4 days, followed by melphalan (HDM: 140 mg/m2) and autologous stem cell transplantation (ASCT). The anti-IL-6 monoclonal antibody (mAb) (B-E8) was given till haematological recovery, starting 1 day before DEX. Results were historically compared to MM patients treated with HDM 140 and 200 mg/m2. Our results show (1) that B-E8 was able to fully neutralize IL-6 activity in vivo before and after HDM as shown by inhibition of C reactive protein (CRP) production; (2) no haematological toxicity; (3) a significant reduction of mucositis and fever; (4) a median event-free survival of 35 months and an overall survival of 68.2% at 5 years with a median follow-up of 72 months; and (5) the overall daily IL-6 production progressively increased on and after 7 days post-HDM, with the increased serum CRP levels. In the 5/24 patients with uncontrolled CRP production, a large IL-6 production was detected (320 microg/day) that could not possibly be neutralized by B-E8. These data show the feasibility to neutralize IL-6 in vivo with anti-IL-6 mAb in the context of HDM.
Collapse
Affiliation(s)
- J-F Rossi
- Service d'Hématologie et d'Oncologie Médicale, CHU Montpellier, avenue du Doyen Giraud, Montpellier Cédex, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Recent studies have identified the importance of proinflammatory mediators in the development and progression of chronic heart failure (CHF). The growing appreciation of the pathophysiologic consequences of sustained expression of proinflammatory mediators in preclinical and clinical CHF models culminated in a series of multicenter clinical trials that used targeted approaches to neutralize tumor necrosis factor in patients with moderate-to-advanced CHF. However, these targeted approaches have resulted in worsening CHF, thereby raising a number of important questions about what role, if any, proinflammatory cytokines play in the pathogenesis of CHF. This review summarizes what has been learned from the negative clinical trials, as well as the potential direction of future research in this area.
Collapse
Affiliation(s)
- Douglas L Mann
- Winters Center for Heart Failure Research, Cardiology Section, Department of Medicine, Michael E. DeBakey Veterans Administration Medical Center and Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
32
|
Parissis JT, Fountoulaki K, Paraskevaidis I, Kremastinos D. Depression in chronic heart failure: novel pathophysiological mechanisms and therapeutic approaches. Expert Opin Investig Drugs 2005; 14:567-77. [PMID: 15926864 DOI: 10.1517/13543784.14.5.567] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Depression is four to five times as common in chronic heart failure (CHF) patients as in the general population, may confer a higher risk of developing CHF in susceptible populations, and is significantly related to higher hospital readmission rates and increased mortality in established CHF. This effect may be mediated via the pathophysiological mechanisms that are shared between CHF and depression, including increased hypothalamic-pituitary-adrenal function, sympathoadrenal hyperactivity, diminished heart-rate variability and excessive pro-inflammatory cytokine activation. Each of these pathways of linkage represents a potential therapeutic target to improve outcome in CHF. This paper reviews the recent investigational observations that clarify the direct effects of antidepressants on immune functions, as well as the indirect effects of anticytokine pharmacological agents on depressive symptoms in CHF. With recent evidence suggesting that selective serotonin re-uptake inhibitors improve survival after myocardial infarction in patients with depression, diagnosis and treatment of this comorbidity may beneficially affect the functional capacity and prognosis of CHF patients.
Collapse
Affiliation(s)
- John T Parissis
- Attikon University Hospital Heart Failure Clinic, Aftocratoros Irakliou 17, 15122, Maroussi, Athens, Greece.
| | | | | | | |
Collapse
|
33
|
von Haehling S, Anker SD. Future prospects of anticytokine therapy in chronic heart failure. Expert Opin Investig Drugs 2005; 14:163-76. [PMID: 15806710 DOI: 10.1517/13543784.14.2.163] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Several lines of evidence suggest that chronic heart failure is a state of chronic inflammation. Indeed, various pro-inflammatory markers, including the cytokines TNF-alpha, and interleukin 6 and 1, are activated in the course of the disease. In chronic heart failure, these substances are frequently induced even before the classical neurohormones angiotensin II and noradrenaline. Although the recently published anti-TNF-alpha trials with etanercept and infliximab have called the beneficial effects of targeting single cytokines into question, the overactive immune system remains a promising target for therapeutic interventions, which aim at slowing down disease progression. Broader approaches are required. These comprise targeting bacterial lipopolysaccharide (endotoxin) that enters the circulation through the oedematous gut wall, immune modulation therapy with patient-derived whole blood exposed to oxidative stress, 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (the so-called statins) and a number of other substances including pentoxifylline and thalidomide.
Collapse
Affiliation(s)
- Stephan von Haehling
- Imperial College School of Medicine, Department of Clinical Cardiology, NationalHeart & Lung Institute, Dovehouse Street, London, SW3 6LY, UK.
| | | |
Collapse
|
34
|
Berthonneche C, Sulpice T, Boucher F, Gouraud L, de Leiris J, O'Connor SE, Herbert JM, Janiak P. New insights into the pathological role of TNF-alpha in early cardiac dysfunction and subsequent heart failure after infarction in rats. Am J Physiol Heart Circ Physiol 2004; 287:H340-50. [PMID: 15210453 DOI: 10.1152/ajpheart.01210.2003] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A marked increase in plasma TNF-alpha has been described in patients with chronic heart failure (CHF). Nevertheless, little is known about the direct role of this cytokine early after myocardial infarction (MI) and its possible effects on the subsequent development of CHF. Wistar rats were subjected to permanent in vivo coronary artery ligation. At 5, 7, and 9 days after MI, cardiac function, passive compliance of the left ventricle (LV), and cardiac geometry were evaluated. The same model was used to perform pharmacological studies 7 days and 10 wk after MI in rats treated with monomeric recombinant human soluble TNF-alpha receptor type II (sTNF-RII, 40 microg/kg iv) or a placebo on day 3. Maximal alterations of cardiac function and geometry occurred 7 days after MI, which correlated chronologically with a peak of cardiac and serum TNF-alpha, as shown by immunohistochemistry and ELISA, respectively. sTNF-RII improved LV end-diastolic pressure under basal conditions and after volume overload 7 days and 10 wk after MI. Moreover, a significant leftward shift of the pressure-volume curve in the sTNF-RII-treated group 7 days after MI indicated a preservation of LV volume. Infarct expansion index was also significantly improved by sTNF-RII 7 days after MI (P < 0.01). Nevertheless, 10 wk after MI, geometric indexes and passive pressure-volume curves were not significantly improved by the treatment. In conclusion, TNF-alpha plays a major role in cardiac alterations 7 days after MI in rats and contributes to hemodynamic derangement, but not to cardiac remodeling, in subsequent CHF.
Collapse
|
35
|
Begg MJ, Sturrock ED, van der Westhuyzen DR. Soluble LDL-R are formed by cell surface cleavage in response to phorbol esters. ACTA ACUST UNITED AC 2004; 271:524-33. [PMID: 14728679 DOI: 10.1046/j.1432-1033.2003.03953.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A 140-kDa soluble form of the low density lipoprotein (LDL) receptor has been isolated from the culture medium of HepG2 cells and a number of other cell types. It is produced from the 160-kDa mature LDL receptor by a proteolytic cleavage, which is stimulated in the presence of 4beta-phorbol 12-myristate 13-acetate (PMA), leading to the release of a soluble fragment that constitutes the bulk of the extracellular domain of the LDL receptor. By labeling HepG2 cells with [35S]methionine and chasing in the presence of PMA, we demonstrated that up to 20% of LDL-receptors were released into the medium in a 2-h period. Simultaneously, the level of labeled cellular receptors was reduced by 30% in those cells treated with PMA compared to untreated cells, as was the total number of cell surface LDL-receptors assayed by the binding of 125I-labeled antibody to whole cells. To determine if endocytosis was required for cleavage, internalization-defective LDL-receptors were created by mutagenesis or deletion of the NPXY internalization signal, transfected into Chinese hamster ovary cells, and assayed for cleavage in the presence and absence of PMA. Cleavage was significantly greater in the case of the mutant receptors than for wild-type receptors, both in the absence and presence of PMA. Similar results were seen in human skin fibroblasts homozygous for each of the internalization-defective LDL receptor phenotypes. LDL receptor cleavage was inhibited by the hydoxamate-based inhibitor TAPI, indicating the resemblance of the LDL receptor cleavage mechanism to that of other surface released membrane proteins.
Collapse
Affiliation(s)
- Michael J Begg
- Division of Medical Biochemistry, University of Cape Town, South Africa
| | | | | |
Collapse
|
36
|
Soos JM, Polsky RM, Keegan SP, Bugelski P, Herzyk DJ. Identification of natural antibodies to interleukin-18 in the sera of normal humans and three nonhuman primate species. Clin Immunol 2003; 109:188-96. [PMID: 14597217 DOI: 10.1016/s1521-6616(03)00180-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Natural antibodies to cytokines can be found in the sera of normal healthy individuals in the absence of specific immunostimulation. However, the function, impact, and purpose of natural antibody development have yet to be fully elucidated. Interleukin (IL)-18 is a cytokine that exerts proinflammatory activities and induces natural killer (NK) cell activity. Recombinant human IL-18 (rHuIL-18) is currently in development as a cancer immunotherapy. In this study, the presence of natural antibodies to IL-18 in the sera of normal humans and three nonhuman primate species was evaluated by electrochemiluminescence immunoassay (ECLIA). Of the human sera tested, 6 of 47 samples were positive for natural antibodies to IL-18. Of the nonhuman primate sera tested, 22 of 80 cynomolgus monkey samples, 4 of 31 rhesus monkey samples, and 2 of 20 chimpanzee samples were positive for natural antibodies to IL-18. Natural anti-IL-18 antibodies were neutralizing in 5 of 22 cynomolgus and 2 of 4 rhesus sera. None of the chimpanzee or human sera were able to neutralize IL-18 induction of interferon (IFN)-gamma in vitro. In vivo activity of rHuIL-18 was compared in IL-18 natural antibody-positive and -negative cynomolgus monkeys. The presence of natural antibodies to IL-18 did not alter rHuIL-18 systemic exposure levels, induction of neopterin, or induction of treatment-induced antibodies following intravenous administration of rHuIL-18. In conclusion, our data indicate that, as has been found with other cytokines, natural anti-IL-18 antibodies are relatively common. Moreover, natural anti-IL-18 antibodies do not appear to influence rHuIL-18 activity in vivo and are not predictive of a heightened immune response, suggesting that natural anti-IL-18 antibodies do not impact IL-18 therapy. Finally, our data suggest that the ability to detect natural anti-cytokine antibodies may be a useful measure of the adequacy of an assay for deployment in clinical trials.
Collapse
Affiliation(s)
- Jeanne M Soos
- Department of Safety Assessment, GlaxoSmithKline Pharmaceuticals, 709 Swedeland Road, UE0368, King of Prussia, PA 19406, USA.
| | | | | | | | | |
Collapse
|
37
|
Abstract
Recent studies have identified the importance of proinflammatory mediators in the development and progression of heart failure. The growing appreciation of the pathophysiological consequences of sustained expression of proinflammatory mediators in preclinical and clinical heart failure models culminated in a series of multicenter clinical trials that used "targeted" approaches to neutralize tumor necrosis factor in patients with moderate to advanced heart failure. However, these targeted approaches have resulted in worsening heart failure, thereby raising a number of important questions about what role, if any, proinflammatory cytokines play in the pathogenesis of heart failure. This review will summarize the tremendous growth of knowledge that has taken place in this field, with a focus on what we have learned from the negative clinical trials, as well as the potential direction of future research in this area.
Collapse
Affiliation(s)
- Douglas L Mann
- Winters Center for Heart Failure Research, the Cardiology Section, Department of Medicine, Veterans Administration Medical Center, Methodist Hospital, and Baylor College of Medicine, Houston, Tex 77030, USA.
| |
Collapse
|
38
|
Beyaert R, Van Loo G, Heyninck K, Vandenabeele P. Signaling to gene activation and cell death by tumor necrosis factor receptors and Fas. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 214:225-72. [PMID: 11893167 DOI: 10.1016/s0074-7696(02)14007-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor (TNF) receptors and Fas elicit a wide range of biological responses, including cell death, cell proliferation, inflammation, and differentiation. The pleiotropic character of these receptors is reflected at the level of signal transduction. The cytotoxic effects of TNF and Fas result from the activation of an apoptotic/necrotic program. On the other hand, TNF receptors, and under certain conditions also Fas, exert a proinflammatory function that results from the induction of several genes. In this context, the transcription factor nuclear factor-kappa B (NF-kappaB) plays an important role. NF-kappaB is also important for the induction of several antiapoptotic genes, which explains at least partially why several cell types can only be killed by TNF in the presence of transcription or translation inhibitors. It is the balance between proapoptotic and antiapoptotic pathways that determines whether a cell will finally die or proliferate. A third signal transduction pathway that is activated in response to TNF is the mitogen-activated protein kinase cascade, which plays an important role in the modulation of transcriptional gene activation.
Collapse
Affiliation(s)
- Rudi Beyaert
- Department of Molecular Biology, University of Gent-Flanders Interuniversity Institute for Biotechnology, Belgium
| | | | | | | |
Collapse
|
39
|
Su SJ, Yeh TM. The dynamic responses of pro-inflammatory and anti-inflammatory cytokines of human mononuclear cells induced by uromodulin. Life Sci 2000; 65:2581-90. [PMID: 10619366 DOI: 10.1016/s0024-3205(99)00527-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
This study was undertaken to examine the dynamic response of human peripheral blood mononuclear cells (PBMC) in the secretion of proinflammatory and anti-inflammatory cytokines induced by uromodulin (URO). Levels of tumor necrosis factor-alpha (TNFalpha), TNF soluble receptor (sTNFRI and II), interleukin 1-beta (IL-1beta), and IL-1 receptor antagonist (IL-1Ra) in the supernatants of URO-stimulated PBMC were measured by ELISA. URO stimulated the secretion of all these cytokines in a dose dependent manner except sTNFRI. Peak levels of TNFalpha and IL-1beta were reached at 6-12 h, while 5-10 fold higher in sTNFR II and IL-1Ra levels were observed at 24-48 h after URO stimulation. URO-induced secretion of TNFalpha, IL-1beta, sTNFRII and IL-1Ra could be enhanced by human plasma. Specifically, serum proteins including C3, sCD14 and IgG not only bound to URO but also enhanced URO-induced TNFalpha secretion of PBMC. Collectively, our data suggest that URO might have dual immunomodulating effect through regulating the secretion of proinflammatory and anti-inflammatory cytokines, and that serum binding proteins might enhance this activity.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Antigens, CD/biosynthesis
- Carrier Proteins/blood
- Female
- Humans
- Interleukin 1 Receptor Antagonist Protein
- Interleukin-1/biosynthesis
- Interleukin-1/blood
- Interleukin-1/metabolism
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Monokines/biosynthesis
- Monokines/blood
- Monokines/metabolism
- Mucoproteins/blood
- Mucoproteins/pharmacology
- Pregnancy
- Pregnancy Proteins/pharmacology
- Receptors, Interleukin-1/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Sialoglycoproteins/biosynthesis
- Sialoglycoproteins/blood
- Sialoglycoproteins/metabolism
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/metabolism
- Uromodulin
Collapse
Affiliation(s)
- S J Su
- Department of Microbiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | |
Collapse
|
40
|
Di Marco R, Xiang M, Zaccone P, Leonardi C, Franco S, Meroni P, Nicoletti F. Concanavalin A-induced hepatitis in mice is prevented by interleukin (IL)-10 and exacerbated by endogenous IL-10 deficiency. Autoimmunity 1999; 31:75-83. [PMID: 10680745 DOI: 10.3109/08916939908994050] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
One single intra-venous (i.v.) injection of Concanavalin A (Con A) into mice provokes a cell-mediated immunoinflammatory hepatitis. We have presently evaluated the immunopharmacological effects of exogenous interleukin (IL)-10 and the role of endogenous IL-10 in this model by using exogenous IL-10, anti-IL-10 monoclonal antibody (mAb) and mice with disrupted IL-10 gene (IL-10 KO mice). Whilst exogenous IL-10 administered in a prophylactic (1 h prior to Con A) and even "early" therapeutic fashion (30 min after Con A) reduced the elevation of transaminase activities in plasma in a dose-dependent manner, observed in control mice, these biochemical markers of liver injury were significantly increased both in IL-10 KO mice as well as in those receiving anti-IL-10 mAb. Interestingly, doses of Con A lower than 20 mg/kg that were only capable of inducing slight serological signs of hepatitis in mice, exerted marked hepatitic effects when administered to either anti-IL-10 mAb-treated mice or to IL-10 KO mice. The disease modulating effects of exogenous IL-10 and either genetical or pharmacologically-induced IL-10 deficiency were associated with profound and opposite modifications of the Con A-induced increase in the circulating levels of IFN-gamma and TNF-alpha. Relative to control animals, the blood levels of these cytokines were diminished in IL-10-treated mice and augmented in both IL-10 KO mice and anti-IL-10 mAb-treated mice. These results prove the physiological antiinflammatory role of endogenous IL-10 in Con A induced hepatitis and the beneficial effects of IL-10 treatment to prevent this condition.
Collapse
Affiliation(s)
- R Di Marco
- Department of Microbiological and Gynaecological Science, Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
Xiang M, Zaccone P, Di Marco R, Harris R, Magro G, Di Mauro M, Meroni PL, Garotta G, Nicoletti F. Failure of exogenously administered interferon-gamma or blockage of endogenous interleukin-4 with specific inhibitors to augment the incidence of autoimmune diabetes in male NOD mice. Autoimmunity 1999; 30:71-80. [PMID: 10435719 DOI: 10.3109/08916939908994763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Interferon (IFN)-gamma and interleukin (IL)-4 are prototypic type 1 and type 2 cytokines which are known to play pathogenetic and protective roles, respectively, in NOD mouse IDDM. The capacity of male NOD mice to produce more IL-4 and less IFN-gamma within the insulitic lesions than females has been suggested to contribute to their lower incidence of diabetes. In this study we have tested the effects of prolonged prophylactic treatment of male NOD mice with rat IFN-gamma, mouse IFN-gamma, anti-IL-4 monoclonal antibody (mAb) and recombinant murine soluble IL-4 receptor (smIL-4R) on the diabetogenic events leading to insulitis and diabetes. None of these treatments influenced spontaneous and/or cyclophosphamide-induced autoimmune diabetogenesis in male NOD mice. Control mice exhibited comparable histological signs of insulitis and incidence of diabetes to those treated with either mouse/rat IFN-gamma or specific IL-4 inhibitors. On the contrary, both clinical and histological signs of diabetes were suppressed by prophylactic treatment with anti-IFN-gamma mAb. These findings indicate that the autoimmune diathesis of male NOD mice towards IDDM cannot be augmented by manipulation of endogenous IFN-gamma or IL-4.
Collapse
Affiliation(s)
- M Xiang
- Institute of Microbiology, University of Catania, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Quattrocchi E, Walmsley M, Browne K, Williams RO, Marinova-Mutafchieva L, Buurman W, Butler DM, Feldmann M. Paradoxical Effects of Adenovirus-Mediated Blockade of TNF Activity in Murine Collagen-Induced Arthritis. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.2.1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Collagen-induced arthritis (CIA) is an experimental model of arthritis widely used to dissect the pathogenesis of human rheumatoid arthritis and to identify potential therapeutic targets. Among these, TNF-α has been recognized to play an important role. Here we investigate the feasibility and therapeutic efficacy of prolonged blockade of TNF-α activity through the adenovirus-mediated gene delivery of a dimeric chimeric human p55 TNFR-IgG fusion protein and compare it to protein therapy in established CIA. A single i.v. administration of the replication-deficient adenovirus yielded microgram serum levels of the chimeric fusion protein and ameliorated CIA for 10 days. Subsequently, benefit was lost and a rebound to greater inflammatory activity was observed despite the continual presence of bioactive TNFR fusion protein. A similar trend was also observed in mice injected directly with comparable amounts of a human TNFR-IgG fusion protein, whereas the administration of a control adenovirus-encoding β-galactosidase or of a control human IgG1 protein did not significantly affect the disease course. The mechanisms of the rebound of CIA were investigated, and augmented Ab response to collagen type II and TNFR were identified as potential causes. Our results confirm the feasibility of adenovirus-mediated gene delivery of cytokine inhibitors in animal models of autoimmune diseases for investigational purposes and highlight the importance of prolonged studies. Further investigations are needed to optimize ways of exploiting the potential of adenoviral gene therapy in RA.
Collapse
Affiliation(s)
| | - Marita Walmsley
- *Kennedy Institute of Rheumatology, London, United Kingdom; and
| | - Kylie Browne
- *Kennedy Institute of Rheumatology, London, United Kingdom; and
| | | | | | - Wim Buurman
- †Department of Surgery, University of Limburg, Maastricht, The Netherlands
| | - Debra M. Butler
- *Kennedy Institute of Rheumatology, London, United Kingdom; and
| | - Marc Feldmann
- *Kennedy Institute of Rheumatology, London, United Kingdom; and
| |
Collapse
|
43
|
Gardiner SM, Kemp PA, March JE, Bennett T. Influence of FR 167653, an inhibitor of TNF-alpha and IL-1, on the cardiovascular responses to chronic infusion of lipopolysaccharide in conscious rats. J Cardiovasc Pharmacol 1999; 34:64-9. [PMID: 10413069 DOI: 10.1097/00005344-199907000-00011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Conscious, male Long Evans rats (350-450 g) chronically instrumented for the measurement of regional haemodynamics, were infused with FR 167653, a dual inhibitor of tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 (IL-1) synthesis (0.32 mg/kg/h) for 24 h, beginning 1 h before coinfusion of saline, or with saline for 24 h beginning 1 h before coinfusion of lipopolysaccharide (150 microg/kg/h), or with FR 167653 beginning 1 h before coinfusion of lipopolysaccharide. Animals infused with FR 167653 and saline showed progressive hindquarters vasoconstriction over the 24-h period, but this was not different from the change seen in animals (n = 3) infused with saline alone. However, plasma analysis at the end of the coinfusion of FR 167653 and saline showed substantial elevation in levels of creatine kinase, lactate dehydrogenase, and potassium, consistent with some tissue damage (heart, liver, or skeletal muscle, or a combination of these). Animals coinfused with saline and lipopolysaccharide showed biphasic decreases in mean arterial blood pressure accompanied by renal hyperaemic vasodilatation, and decreases followed by increases in mesenteric and hindquarters flows and vascular conductances. At the end of the infusion period, plasma analysis showed signs of renal dysfunction (elevated creatinine) and hepatic dysfunction (elevated alkaline phosphatase, gamma-glutamyl transferase, and alanine aminotransferase). In the presence of FR 167653, the hypotensive effects of lipopolysaccharide were abolished, but regional haemodynamics were unchanged, as were signs of organ dysfunction. One explanation of these observations is that FR 167653 causes a relative improvement in cardiac function during infusion of lipopolysaccharide, and this opposes the hypotensive effects of the latter, in spite of its persistent vasodilator effects.
Collapse
Affiliation(s)
- S M Gardiner
- School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, England, UK
| | | | | | | |
Collapse
|
44
|
Klein B, Lu ZY, Gu ZJ, Costes V, Jourdan M, Rossi JF. Interleukin-10 and Gp130 cytokines in human multiple myeloma. Leuk Lymphoma 1999; 34:63-70. [PMID: 10350333 DOI: 10.3109/10428199909083381] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Interleukin (IL)-10 is a critical cytokine involved in the terminal differentiation of B cells into plasma cells. IL-10 is also involved in multiple myeloma, a malignant plasma cell disorder. IL-6 and, more generally the cytokines activating the gp130 IL-6 transducer, are major survival and proliferation factors of myeloma cells. IL-10 is also a growth factor of malignant plasma cells, produced by myeloma cells from about half the patients and is detected in the plasma of patients with plasma cell leukemia or solitary plasmacytoma. The myeloma cell growth activity of IL-10 is mediated through a gp130 cytokine, oncostatin M (OSM), that is frequently produced by myeloma cells. Myeloma cells fail to express OSM receptors but IL-10, by inducing it, confers on them the sensitivity to OSM.
Collapse
Affiliation(s)
- B Klein
- INSERM U475, Montpellier, France.
| | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Borsellino N, Bonavida B, Ciliberto G, Toniatti C, Travali S, D'Alessandro N. Blocking signaling through the gp130 receptor chain by interleukin-6 and oncostatin M inhibits PC-3 cell growth and sensitizes the tumor cells to etoposide and cisplatin-mediated cytotoxicity. Cancer 1999. [DOI: 10.1002/(sici)1097-0142(19990101)85:1<134::aid-cncr19>3.0.co;2-c] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
47
|
Abstract
Cytokines participate in the induction and effector phases of all immune and inflammatory responses. They are therefore obvious tools and targets for strategies designed to promote, inhibit or redirect these responses. However, the complexity of the cytokine network has hindered the widespread clinical application of many cytokines and it has become clear that a deeper understanding of the normal operation of this system in health and disease is needed for the therapeutic potential of cytokines to be fully realized. This review summarizes some of the principles that are now thought to underlie the diverse functions of the interleukins, interferons, colony-stimulating factors and tumour necrosis factors in immune and inflammatory reactions in vivo. Genetic and structural relationships between these cytokines, the regulation of their synthesis, and the structures and functions of their receptors are outlined. Current knowledge of these parameters suggests ways in which multiple positive and negative regulatory mechanisms are integrated to balance cytokine benefits and harm under physiological conditions and offers new prospects for rational exploitation of this system.
Collapse
Affiliation(s)
- A Kelso
- The Queensland Institute of Medical Research, Brisbane, Australia.
| |
Collapse
|
48
|
Bendtzen K, Hansen MB, Ross C, Svenson M. High-avidity autoantibodies to cytokines. IMMUNOLOGY TODAY 1998; 19:209-11. [PMID: 9613037 DOI: 10.1016/s0167-5699(98)01252-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- K Bendtzen
- Institute for Inflammation Research, Rigshospitalet, Copenhagen, Denmark
| | | | | | | |
Collapse
|
49
|
Antibody to Granulocyte-Macrophage Colony-Stimulating Factor Is a Dominant Anti-Cytokine Activity in Human IgG Preparations. Blood 1998. [DOI: 10.1182/blood.v91.6.2054.2054_2054_2061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pharmaceutical preparations of normal human immunoglobulin (IgG) are known to contain high-avidity and neutralizing antibodies (Ab) to the cytokines interleukin (IL)-1α, IL-6, and interferon (IFN)α. To test for other cytokine Ab, 23 batches of IgG were tested for saturable binding to eight 125I-labeled recombinant cytokines. All batches bound granulocyte-macrophage colony-stimulating factor (GM-CSF) with high avidity (Kav ≈ 10 pmol/L) and capacities of up to 5 μmol GM-CSF/mol IgG. Only 1 of 15 batches bound IL-5, also with high avidity, whereas 13 of 15 batches bound to IL-10 but with lower capacities and avidities. None of the IgG preparations bound IL-1 receptor antagonist (IL-1ra), IL-2, IL-3, IL-4, or G-CSF. Cross-binding and absorption analyses revealed identical or slightly stronger binding of recombinant GM-CSF, IL-5, and IL-10 than their native counterparts. GM-CSF–IgG complexes did not bind to cellular GM-CSF receptors, but Fc-dependent binding occurred to blood polymorphonuclear cells. Increased binding of GM-CSF to patient sera correlated positively with the binding capacities of infused IgG preparations. Patient and normal sera did not interfere with the binding of Ab to GM-CSF. From these and previous experiments, we conclude that pools of normal human IgG contain variable amounts of specific and high-avidity Ab to some cytokines, and that Ab to GM-CSF constitute a dominant anti-cytokine activity in these preparations. These Ab are available for reactionin vivo following IgG therapy.
Collapse
|
50
|
Antibody to Granulocyte-Macrophage Colony-Stimulating Factor Is a Dominant Anti-Cytokine Activity in Human IgG Preparations. Blood 1998. [DOI: 10.1182/blood.v91.6.2054] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Pharmaceutical preparations of normal human immunoglobulin (IgG) are known to contain high-avidity and neutralizing antibodies (Ab) to the cytokines interleukin (IL)-1α, IL-6, and interferon (IFN)α. To test for other cytokine Ab, 23 batches of IgG were tested for saturable binding to eight 125I-labeled recombinant cytokines. All batches bound granulocyte-macrophage colony-stimulating factor (GM-CSF) with high avidity (Kav ≈ 10 pmol/L) and capacities of up to 5 μmol GM-CSF/mol IgG. Only 1 of 15 batches bound IL-5, also with high avidity, whereas 13 of 15 batches bound to IL-10 but with lower capacities and avidities. None of the IgG preparations bound IL-1 receptor antagonist (IL-1ra), IL-2, IL-3, IL-4, or G-CSF. Cross-binding and absorption analyses revealed identical or slightly stronger binding of recombinant GM-CSF, IL-5, and IL-10 than their native counterparts. GM-CSF–IgG complexes did not bind to cellular GM-CSF receptors, but Fc-dependent binding occurred to blood polymorphonuclear cells. Increased binding of GM-CSF to patient sera correlated positively with the binding capacities of infused IgG preparations. Patient and normal sera did not interfere with the binding of Ab to GM-CSF. From these and previous experiments, we conclude that pools of normal human IgG contain variable amounts of specific and high-avidity Ab to some cytokines, and that Ab to GM-CSF constitute a dominant anti-cytokine activity in these preparations. These Ab are available for reactionin vivo following IgG therapy.
Collapse
|