1
|
Dong H, Wang T, Wang M, Yan Y, Zhang X, Gu W, Ji W, Huang L, Chen Z. The role of inducible costimulatory molecular ligand (ICOSL) in children with neutrophilic asthma. Transl Pediatr 2020; 9:469-479. [PMID: 32953544 PMCID: PMC7475312 DOI: 10.21037/tp-20-172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND It has been shown that certain severe and refractory asthma cases are caused by neutrophil and not eosinophil infiltration. Inducible costimulatory molecular ligand (ICOSL) expression is closely associated with tumor and autoimmune diseases, yet a limited amount of data has been published regarding the significance of ICOSL in children with neutrophilic asthma. The present study aimed to explore the clinical significance of abnormal expression of ICOSL in peripheral blood and bronchoalveolar lavage fluid (BALF) samples of children with neutrophilic asthma. METHODS Selected children from the Children's Hospital of Soochow University who met the diagnostic criteria of asthma and excluded patients with a pathogen-positive etiology. Children who were admitted to the hospital for foreign body inhalation in the same period acted as the control group. Children with more than 50% of neutrophils in BALF samples were assigned to the neutrophilic asthma group (NA group), and the remaining subjects composed the asthma group (A group). The expression levels of ICOSL, IL-4, IL-17, IFN-γ, neutrophil elastase (NE), and matrix metalloproteinase-9 (MMP-9) were detected in plasma and BALF samples by enzyme-linked immunosorbent assays, in order to analyze the differences in the levels of cytokines and clinical characteristics between children with neutrophilic asthma and non-neutrophilic asthma. Moreover, the potential mechanism of ICOSL in neutrophilic asthma was explored. RESULTS 32 children were enrolled: 12 children in the NA group and 20 children in the A group. The mean hospitalization time of the NA group was longer than that of the A group (P<0.05). The concentration levels of ICOSL, IL-17, NE, and MMP-9 in plasma and BALF samples in the NA group were higher than those in the A group, while the levels of IFN-γ exhibited opposite. A significant correlation was found between ICOSL and IL-17 levels in plasma (r=0.753, P=0.012) and BALF (r=0.774, P=0.009) samples in the NA group. CONCLUSIONS Children with neutrophilic asthma were more severely affected, experiencing a considerably more difficult clinical treatment and longer hospitalization time. ICOSL may regulate the secretion of IL-17 by Th17 and increase the levels of NE and MMP-9, which are involved in the development of immune inflammation in neutrophils.
Collapse
Affiliation(s)
- Heting Dong
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Ting Wang
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Meijuan Wang
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Yongdong Yan
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Xinxing Zhang
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Wenjing Gu
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Wei Ji
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Li Huang
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Zhengrong Chen
- Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
2
|
Mohammadi M, Dehghani P, Mohseninia A, Roozbehani M, Hemphill A, Hesamizadeh K. Incorporation of the Tat cell-penetrating peptide into nanofibers improves the respective antitumor immune response. J Cell Physiol 2020; 236:1401-1417. [PMID: 32686113 DOI: 10.1002/jcp.29946] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/07/2020] [Indexed: 11/06/2022]
Abstract
A major challenge for the development of anticancer vaccines is the induction of a safe and effective immune response, particularly mediated by CD8+ T lymphocytes, in an adjuvant-free manner. In this respect, we present a simple strategy to improve the specific CD8+ T cell responses using KFE8 nanofibers bearing a Class I (Kb)-restricted peptide epitope (called E. nanofibers) without the use of adjuvant. We demonstrate that incorporation of Tat, a cell-penetrating peptide (CPP) of the HIV transactivator protein, into E. nanofibers remarkably enhanced tumor-specific CD8+ T cell responses. E. nanofibers containing 12.5% Tat peptide (E.Tat12.5 nanofiber) increased antigen cross-presentation by bone marrow-derived dendritic cells as compared with E. nanofibers, or E. nanofibers containing 25 or 50% the Tat peptide. Uptake of KFE8.Tat12.5 nanofibers by dendritic cells (DCs) was significantly increased compared with KFE8 nanofiber lacking Tat. Peritoneal and lymph node DCs of mice immunized with E.Tat12.5 nanofibers exhibited increased presentation of the H2kb-epitope (reminiscent for cross-presentation) compared with DCs obtained from E. nanofiber vaccinated mice. Tetrameric and intracellular cytokine staining revealed that vaccination with E.Tat12.5 triggered a robust and specific CD8+ T lymphocyte response, which was more pronounced than in mice vaccinated with E. nanofibers alone. Furthermore, E.Tat12.5 nanofibers were more potent than E. nanofiber to induce antitumor immune response and tumor-infiltrating IFN-γ CD8 T lymphocyte. In terms of cancer vaccine development, we propose that harnessing the nanofiber-based vaccine platform with incorporated Tat peptide could present a simple and promising strategy to induce highly effective antitumor immune response.
Collapse
Affiliation(s)
- Mohsen Mohammadi
- Biotechnology Department, The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Parva Dehghani
- Biotechnology Department, The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Atefeh Mohseninia
- Department of Biochemistry, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mona Roozbehani
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Andrew Hemphill
- Department of Infectious Diseases and Pthobiology, Institute of Parasitology, Vetsuisse Faculty, University of Berne, Bern, Switzerland
| | - Khashayar Hesamizadeh
- Department of Virology, Middle East Liver Diseases (MELD) Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
He W, Wang B, Li Q, Yao Q, Jia X, Song R, Li S, Zhang JA. Aberrant Expressions of Co-stimulatory and Co-inhibitory Molecules in Autoimmune Diseases. Front Immunol 2019; 10:261. [PMID: 30842773 PMCID: PMC6391512 DOI: 10.3389/fimmu.2019.00261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/29/2019] [Indexed: 12/26/2022] Open
Abstract
Co-signaling molecules include co-stimulatory and co-inhibitory molecules and play important roles in modulating immune responses. The roles of co-signaling molecules in autoimmune diseases have not been clearly defined. We assessed the expressions of co-stimulatory and co-inhibitory molecules in autoimmune diseases through a bioinformatics-based study. By using datasets of whole-genome transcriptome, the expressions of 54 co-stimulatory or co-inhibitory genes in common autoimmune diseases were analyzed using Robust rank aggregation (RRA) method. Nineteen array datasets and 6 RNA-seq datasets were included in the RRA discovery study and RRA validation study, respectively. Significant genes were further validated in several autoimmune diseases including Graves' disease (GD). RRA discovery study suggested that CD160 was the most significant gene aberrantly expressed in autoimmune diseases (Adjusted P = 5.9E-12), followed by CD58 (Adjusted P = 5.7E-06) and CD244 (Adjusted P = 9.5E-05). RRA validation study also identified CD160 as the most significant gene aberrantly expressed in autoimmune diseases (Adjusted P = 5.9E-09). We further found that the aberrant expression of CD160 was statistically significant in multiple autoimmune diseases including GD (P < 0.05), and CD160 had a moderate role in diagnosing those autoimmune diseases. Flow cytometry confirmed that CD160 was differentially expressed on the surface of CD8+ T cells between GD patients and healthy controls (P = 0.002), which proved the aberrant expression of CD160 in GD at the protein level. This study suggests that CD160 is the most significant co-signaling gene aberrantly expressed in autoimmune diseases. Treatment strategy targeting CD160-related pathway may be promising for the therapy of autoimmune diseases.
Collapse
Affiliation(s)
- Weiwei He
- Department of Endocrinology, Affiliated Hospital of Yanan Medical University, Yanan, China
| | - Bin Wang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Qian Li
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Qiuming Yao
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xi Jia
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Ronghua Song
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Sheli Li
- Department of Endocrinology, Affiliated Hospital of Yanan Medical University, Yanan, China
| | - Jin-An Zhang
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
4
|
|
5
|
The Costimulatory Pathways and T Regulatory Cells in Ischemia-Reperfusion Injury: A Strong Arm in the Inflammatory Response? Int J Mol Sci 2018; 19:ijms19051283. [PMID: 29693595 PMCID: PMC5983665 DOI: 10.3390/ijms19051283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 04/14/2018] [Accepted: 04/19/2018] [Indexed: 02/08/2023] Open
Abstract
Costimulatory molecules have been identified as crucial regulators in the inflammatory response in various immunologic disease models. These molecules are classified into four different families depending on their structure. Here, we will focus on various ischemia studies that use costimulatory molecules as a target to reduce the inherent inflammatory status. Furthermore, we will discuss the relevant role of T regulatory cells in these inflammatory mechanisms and the costimulatory pathways in which they are involved.
Collapse
|
6
|
Wang M, Zhou G, Lv J, Zeng P, Guo C, Wang Q. Curcumin modulation of the activation of PYK2 in peripheral blood mononuclear cells from patients with lupus nephritis. Reumatologia 2017; 55:269-275. [PMID: 29491534 PMCID: PMC5825964 DOI: 10.5114/reum.2017.72623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/01/2017] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Proline-rich tyrosine kinase 2 (PYK2) provides important signals during the activation of lymphocytes, which is essential in autoimmune diseases. Systemic lupus erythematosus (SLE) is a representative autoimmune disease, and lupus nephritis (LN) is one of its most severe complications. Although glucocorticoid-binding immuno-suppression is the first-line therapy for patients with LN, the common and severe side effects of such treatment call for new strategies to improve long-term prognosis and life quality for these patients. Curcumin has been used to treat autoimmune disease with good curative effect, but little is known about the effect of curcumin on LN patients. Our aim was to investigate the mechanism of curcumin for management of LN, specifically regarding the PYK2 pathways. MATERIAL AND METHODS Freshly isolated peripheral blood mononuclear cells (PBMCs) from 20 LN patients and 20 healthy individuals were cultured and stimulated with either PMA, PMA+TyrA9 (PYK2 specific inhibitor), or PMA+Curcumin, and with PBS as control. After 48 hours of incubation, cells were harvested and the expression of PYK2, p-PYK2, CD40L, CTLA-4, and PBMCs proliferation were measured. Then the expression and activation of PYK2 was evaluated using Western blot, the expression of costimulatory molecules CD40L and CTLA-4 protein was evaluated using flow cytometry, and PBMC proliferation was assessed using a [3H]-thymidine incorporation assay. RESULTS Curcumin inhibited the expression and activation of PYK2 in PBMCs in patients with LN in vitro. The inhibition rate of curcumin was negatively correlated with the level of serum complement, but positively correlated with 24-h proteinuria. Curcumin also suppressed the expression of costimulatory molecules CD40L and CTLA-4, as well as PBMC proliferation. Interestingly, these effects were not reproduced on PBMC cultures of healthy subjects. CONCLUSIONS The inhibition of PYK2 signalling protein may be one of the mechanisms underlying the action of curcumin in LN treatment.
Collapse
Affiliation(s)
- Meiying Wang
- Department of Rheumatology and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Rheumatology and Immunology, The People’s Hospital of Bao’an District, Shenzhen City, China
| | - Gengmin Zhou
- Department of Rheumatology and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jiyang Lv
- Department of Rheumatology and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peiying Zeng
- Department of Rheumatology and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chengshan Guo
- Department of Rheumatology and Immunology, The People’s Hospital of Bao’an District, Shenzhen City, China
| | - Qingwen Wang
- Department of Rheumatology and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
7
|
Vanhove B, Poirier N, Fakhouri F, Laurent L, 't Hart B, Papotto PH, Rizzo LV, Zaitsu M, Issa F, Wood K, Soulillou JP, Blancho G. Antagonist Anti-CD28 Therapeutics for the Treatment of Autoimmune Disorders. Antibodies (Basel) 2017; 6:antib6040019. [PMID: 31548534 PMCID: PMC6698823 DOI: 10.3390/antib6040019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/16/2017] [Accepted: 11/18/2017] [Indexed: 12/17/2022] Open
Abstract
The effector functions of T lymphocytes are responsible for most autoimmune disorders and act by directly damaging tissues or by indirectly promoting inflammation and antibody responses. Co-stimulatory and co-inhibitory T cell receptor molecules are the primary pharmacological targets that enable interference with immune-mediated diseases. Among these, selective CD28 antagonists have drawn special interest, since they tip the co-stimulation/co-inhibition balance towards efficiently inhibiting effector T cells while promoting suppression by pre-existing regulatory T-cells. After having demonstrated outstanding therapeutic efficacy in multiple models of autoimmunity, inflammation and transplantation, and safety in phase-I studies in humans, selective CD28 antagonists are currently in early clinical development for the treatment of systemic lupus erythematous and rheumatoid arthritis. Here, we review the available proof of concept studies for CD28 antagonists in autoimmunity, with a special focus on the mechanisms of action.
Collapse
Affiliation(s)
- Bernard Vanhove
- OSE Immunotherapeutics, 44200 Nantes, France.
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Nicolas Poirier
- OSE Immunotherapeutics, 44200 Nantes, France.
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Fadi Fakhouri
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Laetitia Laurent
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
| | - Bert 't Hart
- Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands.
- Department Neuroscience, University of Groningen, University Medical Center, 9713 GZ Groningen, The Netherlands.
| | - Pedro H Papotto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal.
| | - Luiz V Rizzo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil.
| | - Masaaki Zaitsu
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Kathryn Wood
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
| | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| |
Collapse
|
8
|
Ceccarelli F, Perricone C, Cipriano E, Massaro L, Natalucci F, Capalbo G, Leccese I, Bogdanos D, Spinelli FR, Alessandri C, Valesini G, Conti F. Joint involvement in systemic lupus erythematosus: From pathogenesis to clinical assessment. Semin Arthritis Rheum 2017; 47:53-64. [DOI: 10.1016/j.semarthrit.2017.03.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 03/22/2017] [Accepted: 03/31/2017] [Indexed: 12/13/2022]
|
9
|
Yu T, Enioutina EY, Brunner HI, Vinks AA, Sherwin CM. Clinical Pharmacokinetics and Pharmacodynamics of Biologic Therapeutics for Treatment of Systemic Lupus Erythematosus. Clin Pharmacokinet 2017; 56:107-125. [PMID: 27384528 PMCID: PMC5575762 DOI: 10.1007/s40262-016-0426-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease with potentially severe clinical manifestation that mainly affects women of child-bearing age. Patients who do not respond to standard-of-care therapies, such as corticosteroids and immunosuppressants, require biologic therapeutics that specifically target a single or multiple SLE pathogenesis pathways. This review summarizes the clinical pharmacokinetic and pharmacodynamic characteristics of biologic agents that are approved, used off-label, or in the active pipeline of drug development for SLE patients. Depending on the type of target, the interacting biologics may exhibit linear (non-specific) or non-linear (target-mediated) disposition profiles, with terminal half-lives varying from approximately 1 week to 1 month. Biologics given by subcutaneous administration, which offers dosing flexibility over intravenous administration, demonstrated a relatively slow absorption with a time to maximum concentration of approximately 1 day to 2 weeks and a variable bioavailability of 30-82 %. The population pharmacokinetics of monoclonal antibodies were best described by a two-compartment model with central clearance and steady-state volume of distribution ranging from 0.176 to 0.215 L/day and 3.60-5.29 L, respectively. The between-subject variability in pharmacokinetic parameters were moderate (20-79 %) and could be partially explained by body size. The development of linked pharmacokinetic-pharmacodynamic models incorporating SLE disease biomarkers are an attractive strategy for use in dosing regimen simulation and optimization. The relationship between efficacy/adverse events and biologic concentration should be evaluated to improve clinical trial outcomes, especially for biologics in the advanced phase of drug development. New strategies, such as model-based precision dosing dashboards, could be utilized to incorporate information collected from therapeutic drug monitoring into pharmacokinetic/pharmacodynamic models to enable individualized dosing in real time.
Collapse
Affiliation(s)
- Tian Yu
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah School of Medicine, SLC, Utah 295 Chipeta Way, Salt Lake City, UT, 84108, USA
| | - Elena Y Enioutina
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah School of Medicine, SLC, Utah 295 Chipeta Way, Salt Lake City, UT, 84108, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hermine I Brunner
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Catherine M Sherwin
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah School of Medicine, SLC, Utah 295 Chipeta Way, Salt Lake City, UT, 84108, USA.
- Department of Pediatrics, Clinical Trials Office, Salt Lake City, UT, USA.
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
10
|
Mahieu MA, Strand V, Simon LS, Lipsky PE, Ramsey-Goldman R. A critical review of clinical trials in systemic lupus erythematosus. Lupus 2016; 25:1122-40. [PMID: 27497257 PMCID: PMC4978143 DOI: 10.1177/0961203316652492] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
One challenge in caring for patients with systemic lupus erythematosus (SLE) is a paucity of approved therapeutics for treatment of the diverse disease manifestations. In the last 60 years, only one drug, belimumab, has been approved for SLE treatment. Critical evaluation of investigator initiated and pharma-sponsored randomized controlled trials (RCTs) highlights barriers to successful drug development in SLE, including disease heterogeneity, inadequate trial size or duration, insufficient dose finding before initiation of large trials, handling of background medications, and choice of primary endpoint. Herein we examine lessons learned from landmark SLE RCTs and subsequent advances in trial design, as well as discuss efforts to address limitations in current SLE outcome measures that will improve detection of true therapeutic responses in future RCTs.
Collapse
Affiliation(s)
- M A Mahieu
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - V Strand
- Division of Immunology/Rheumatology, Stanford University School of Medicine, Palo Alto, USA
| | | | - P E Lipsky
- AMPEL BioSolutions, Charlottesville, USA
| | - R Ramsey-Goldman
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| |
Collapse
|
11
|
Jansen MF, Hollander MR, van Royen N, Horrevoets AJ, Lutgens E. CD40 in coronary artery disease: a matter of macrophages? Basic Res Cardiol 2016; 111:38. [PMID: 27146510 PMCID: PMC4856717 DOI: 10.1007/s00395-016-0554-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/05/2016] [Indexed: 12/20/2022]
Abstract
Coronary artery disease (CAD), also known as ischemic heart disease (IHD), is the leading cause of mortality in the western world, with developing countries showing a similar trend. With the increased understanding of the role of the immune system and inflammation in coronary artery disease, it was shown that macrophages play a major role in this disease. Costimulatory molecules are important regulators of inflammation, and especially, the CD40L-CD40 axis is of importance in the pathogenesis of cardiovascular disease. Although it was shown that CD40 can mediate macrophage function, its exact role in macrophage biology has not gained much attention in cardiovascular disease. Therefore, the goal of this review is to give an overview on the role of macrophage-specific CD40 in cardiovascular disease, with a focus on coronary artery disease. We will discuss the function of CD40 on the macrophage and its (proposed) role in the reduction of atherosclerosis, the reduction of neointima formation, and the stimulation of arteriogenesis.
Collapse
Affiliation(s)
- Matthijs F Jansen
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Medical Biochemistry, Academic Medical Centre, Meibergdreef 15, 1105AZ, Amsterdam, The Netherlands
| | - Maurits R Hollander
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Niels van Royen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Anton J Horrevoets
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Centre, Meibergdreef 15, 1105AZ, Amsterdam, The Netherlands.
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany.
| |
Collapse
|
12
|
Liu D, Burd EM, Coopersmith CM, Ford ML. Retrogenic ICOS Expression Increases Differentiation of KLRG-1hiCD127loCD8+ T Cells during Listeria Infection and Diminishes Recall Responses. THE JOURNAL OF IMMUNOLOGY 2016; 196:1000-12. [PMID: 26729800 DOI: 10.4049/jimmunol.1500218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 12/02/2015] [Indexed: 12/26/2022]
Abstract
Following T cell encounter with Ag, multiple signals are integrated to collectively induce distinct differentiation programs within Ag-specific CD8(+) T cell populations. Several factors contribute to these cell fate decisions, including the amount and duration of Ag, exposure to inflammatory cytokines, and degree of ligation of cosignaling molecules. The ICOS is not expressed on resting T cells but is rapidly upregulated upon encounter with Ag. However, the impact of ICOS signaling on programmed differentiation is not well understood. In this study, we therefore sought to determine the role of ICOS signaling on CD8(+) T cell programmed differentiation. Through the creation of novel ICOS retrogenic Ag-specific TCR-transgenic CD8(+) T cells, we interrogated the phenotype, functionality, and recall potential of CD8(+) T cells that receive early and sustained ICOS signaling during Ag exposure. Our results reveal that these ICOS signals critically impacted cell fate decisions of Ag-specific CD8(+) T cells, resulting in increased frequencies of KLRG-1(hi)CD127(lo) cells, altered BLIMP-1, T-bet, and eomesodermin expression, and increased cytolytic capacity as compared with empty vector controls. Interestingly, however, ICOS retrogenic CD8(+) T cells also preferentially homed to nonlymphoid organs and exhibited reduced multicytokine functionality and reduced ability to mount secondary recall responses upon challenge in vivo. In sum, our results suggest that an altered differentiation program is induced following early and sustained ICOS expression, resulting in the generation of more cytolyticly potent, terminally differentiated effectors that possess limited capacity for recall response.
Collapse
Affiliation(s)
- Danya Liu
- Emory Transplant Center, Emory University, Atlanta, GA 30322; Department of Surgery, Emory University, Atlanta, GA 30322
| | - Eileen M Burd
- Department of Pathology, Emory University, Atlanta, GA 30322; and
| | - Craig M Coopersmith
- Department of Surgery, Emory University, Atlanta, GA 30322; Emory Critical Care Center, Emory University, Atlanta, GA 30322
| | - Mandy L Ford
- Emory Transplant Center, Emory University, Atlanta, GA 30322;
| |
Collapse
|
13
|
TIGIT negatively regulates inflammation by altering macrophage phenotype. Immunobiology 2016; 221:48-55. [DOI: 10.1016/j.imbio.2015.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/22/2015] [Accepted: 08/11/2015] [Indexed: 11/17/2022]
|
14
|
Snook JD, Chesson CB, Peniche AG, Dann SM, Paulucci A, Pinchuk IV, Rudra JS. Peptide nanofiber–CaCO3 composite microparticles as adjuvant-free oral vaccine delivery vehicles. J Mater Chem B 2016; 4:1640-1649. [DOI: 10.1039/c5tb01623a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
To combat mucosal pathogens that cause gastrointestinal (GI) infections, local mucosal immunity is required which is best achieved through oral vaccination.
Collapse
Affiliation(s)
- Joshua D. Snook
- Department of Pharmacology & Toxicology
- University of Texas Medical Branch
- Galveston
- USA
| | - Charles B. Chesson
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Sealy Center for Vaccine Development
| | - Alex G. Peniche
- Department of Internal Medicine-Division of Infectious Diseases
- University of Texas Medical Branch
- Galveston
- USA
| | - Sara M. Dann
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Department of Internal Medicine-Division of Infectious Diseases
| | | | - Iryna V. Pinchuk
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Department of Internal Medicine-Division of Gastroenterology
| | - Jai S. Rudra
- Department of Pharmacology & Toxicology
- University of Texas Medical Branch
- Galveston
- USA
- Sealy Center for Vaccine Development
| |
Collapse
|
15
|
Luo Y, Yang Y, Zhang H, Zhang T, Wang Y, Tan S, Xu Y, Li D, Ye L, Chen P. Effect of Inducible Co-Stimulatory Molecule siRNA in Cerebral Infarction Rat Models. Med Sci Monit 2015; 21:3003-7. [PMID: 26436531 PMCID: PMC4599179 DOI: 10.12659/msm.894477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background T cell-induced inflammatory response and related cytokine secretion at the injury site may participate in the pathogenesis of cerebral infarction. Recent studies established inducible co-stimulatory molecule (ICOS) as a novel T cell-related factor for its activation and functions. We thus investigate the role of ICOS in cerebral infarction. Material/Methods The siRNA of ICOS was first used to suppress the gene expression in cultured lymphocytes. An in vivo study was then performed by intravenous application of ICOS siRNA in cerebral infarction rats. Survival rates, neurological scores, serum tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-17 levels were observed. Results The expression of ICOS in cultured lymphocytes was significantly suppressed by siRNA. In the in vivo study, the application of siRNA effectively lowered mortality rates of rats, in addition to the improvement of neurological behaviors and amelioration of cerebral tissue damage. Serum levels of TNF-α, IL-1 and IL-17 were all significantly suppressed after siRNA injection. Conclusions ICOS siRNA can protect brain tissues from ischemia injuries after cerebral infarction, improve limb movement and coordination, lower the mortality rate of rats, and inhibit T cell-induced cytokines. These results collectively suggest the potential treatment efficacy of ICOS siRNA against cerebral infarction.
Collapse
Affiliation(s)
- Yingquan Luo
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Yu Yang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Hui Zhang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Ting Zhang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Yina Wang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Shengyu Tan
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Yan Xu
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Dan Li
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Ling Ye
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
16
|
Valor L, López-Longo FJ. [Modulating the survival and maturation system of B lymphocytes: Current and future new therapeutic strategies in systemic lupus erythematosus]. Med Clin (Barc) 2015; 145:206-10. [PMID: 25433780 DOI: 10.1016/j.medcli.2014.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/30/2014] [Accepted: 08/19/2014] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease associated with an aberrant production of autoantibodies by self-reactive B lymphocytes. The study of the phenotypic characteristics of B lymphocytes and the identification of their surface receptors such as BAFF-R, TACI and BCMA, which are responsible of their survival and maturation, have contributed to the development of new therapeutic strategies in recent years.
Collapse
Affiliation(s)
- Lara Valor
- Servicio de Reumatología, Hospital General Universitario Gregorio Marañón, Madrid, España.
| | | |
Collapse
|
17
|
Maurer B, Bosanac I, Shia S, Kwong M, Corpuz R, Vandlen R, Schmidt K, Eigenbrot C. Structural basis of the broadly neutralizing anti-interferon-α antibody rontalizumab. Protein Sci 2015; 24:1440-50. [PMID: 26099203 DOI: 10.1002/pro.2729] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/17/2015] [Indexed: 11/08/2022]
Abstract
Interferons-alpha (IFN-α) are the expressed gene products comprising thirteen type I interferons with protein pairwise sequence similarities in the 77-96% range. Three other widely expressed human type I interferons, IFN-β, IFN-κ and IFN-ω have sequences 29-33%, 29-32% and 56-60% similar to the IFN-αs, respectively. Type I interferons act on immune cells by producing subtly different immune-modulatory effects upon binding to the extracellular domains of a heterodimeric cell-surface receptor composed of IFNAR1 and IFNAR2, most notably anti-viral effects. IFN-α has been used to treat infection by hepatitis-virus type C (HCV) and a correlation between hyperactivity of IFN-α-induced signaling and systemic lupus erythematosis (SLE), or lupus, has been noted. Anti-IFN-α antibodies including rontalizumab have been under clinical study for the treatment of lupus. To better understand the rontalizumab mechanism of action and specificity, we determined the X-ray crystal structure of the Fab fragment of rontalizumab bound to human IFN-α2 at 3Å resolution and find substantial overlap of the antibody and IFNA2 epitopes on IFN-α2.
Collapse
Affiliation(s)
- Brigitte Maurer
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| | - Ivan Bosanac
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| | - Steven Shia
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| | - Mandy Kwong
- Department of Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| | - Racquel Corpuz
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| | - Richard Vandlen
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| | - Kerstin Schmidt
- Department of Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| | - Charles Eigenbrot
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080.,Department of Antibody Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, California, 94080
| |
Collapse
|
18
|
Abstract
As our understanding of the pathogenesis of autoimmune diseases is growing, new therapies are being developed to target disease-specific pathways. Since the introduction of etanercept in 1998, several biotechnological agents have been developed, most of them indicated in the treatment of rheumatoid arthritis, but also psoriatic arthritis. Most currently available molecules target TNF-alfa with different strategies (i.e., etanercept, infliximab, adalimumab, golimumab, and certolizumab pegol), IL-6 (tocilizumab), CTLA-4 (abatacept), and B cells (rituximab, belimumab) as they are key mediators in the cascade of inflammation. Further, small molecules have been recently developed to target intracellular signaling, such as Janus Kinases for tofacitinib, the first FDA-approved small molecule for rheumatoid arthritis. Most novel treatments are being developed for arthritis with specific differences between rheumatoid and psoriatic arthritis, as well as for systemic lupus erythematosus, following the approval of belimumab. Finally, biologic therapies are effective also in gout, mainly targeting interleukin-1 to block the inflammasome. This review article describes the new and upcoming treatment options for rheumatoid arthritis, psoriatic arthritis, systemic lupus erythematosus, and gout to dissect what we should be aware of when discussing these new and promising molecules.
Collapse
|
19
|
Ling C, Liu X, Shen Y, Chen Z, Fan J, Jiang Y, Meng Q. Urinary CD80 levels as a diagnostic biomarker of minimal change disease. Pediatr Nephrol 2015; 30:309-16. [PMID: 25142334 DOI: 10.1007/s00467-014-2915-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/16/2014] [Accepted: 07/14/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Early diagnosis of minimal change disease (MCD) in nephrotic syndrome (NS) patients remains challenging. Doctors often make a diagnosis of MCD using invasive renal biopsy. CD80, a transmembrane protein, is present on podocytes in a number of experimental models of NS. Urinary CD80 levels are significantly elevated in MCD but not in focal segmental glomerulosclerosis (FSGS) or other glomerulopathies. The purpose of this study was to investigate the feasibility of using urinary CD80 levels as a biomarker for the diagnosis of MCD. MATERIALS AND METHODS A total of 165 subjects, 129 men and 36 women, were enrolled in this study. Urinary samples were collected from 37 patients with MCD, 27 patients with FSGS, 30 patients with other glomerulopathies, and 71 healthy people. Using ELISA, experimental values were compared with those produced by kidney biopsy samples. RESULTS The concentration of urinary CD80 was significantly higher in the active MCD group (689.66 ± 378.21 ng/g creatinine) than in the FSGS group (123.49 ± 167. 88 ng/g creatinine, P < 0.00), other glomerulopathies group (152.37 ± 220. 14 ng/g creatinine, P < 0.001) and the control group (81.83 ± 23.01 ng/g creatinine; P < 0.001). A cutoff value of 328.98 (ng/g creatinine) was proposed, with a sensitivity of 81.1 % and specificity of 94.4 %. The area under the receiver operating characteristic (ROC) curve for the urinary CD80 to diagnose MCD was 0.925 (95 % confidence interval: 0.873-0.978). CONCLUSIONS This experiment has preliminarily confirmed urinary CD80 as a non-invasive diagnostic biomarker. It may have significant value in the diagnosis of MCD.
Collapse
Affiliation(s)
- Chen Ling
- Department of Nephrology, Beijing Children's Hospital affiliated to Capital Medical University, West District Nan Li Shi Lu 56th, Beijing, 100045, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Pathogenesis and potential therapeutic targets in systemic lupus erythematosus: from bench to bedside. AUTOIMMUNITY HIGHLIGHTS 2014; 5:33-45. [PMID: 26000154 PMCID: PMC4389042 DOI: 10.1007/s13317-014-0058-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 06/12/2014] [Indexed: 12/18/2022]
Abstract
Systemic lupus erythematosus (SLE) is considered an autoimmune disease with multiorgan involvement. Many advances have been made during the last decade regarding inflammatory pathways, genetic and epigenetic alterations, adaptive and innate immune system mechanisms specifically involved in SLE pathogenesis. Apoptosis has been proposed as an important player in SLE pathogenesis more than a decade ago. However, only recently new key apoptotic pathways have been investigated and the link between apoptotic debris containing autoantigens, innate immunity and ongoing inflammation has been further elucidated. Better understanding of cellular mechanisms and involved cytokines contributed to the development of new biological drugs specifically addressed for SLE therapy.
Collapse
|
21
|
Hoi A, Littlejohn G. Is there still a role for abatacept in the treatment of lupus? Expert Opin Biol Ther 2014; 14:1345-50. [DOI: 10.1517/14712598.2014.935329] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Jüptner M, Zeuner R, Schreiber S, Laudes M, Schröder JO. Successful application of belimumab in two patients with systemic lupus erythematosus experiencing a flare during tocilizumab treatment. Lupus 2014; 23:428-30. [PMID: 24482144 DOI: 10.1177/0961203314520844] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This case report describes two female lupus patients who both received biological treatment with tocilizumab and with belimumab. The disease course was remarkably similar in both cases. Tocilizumab resulted in a transient improvement in pleurisy and arthritis but was then followed by a clinical flare accompanied by an increase in autoantibodies and a drop in complement levels. Alike, both patients experienced a rapid and sustained improvement after institution of belimumab. The clinical benefit obtained is currently stable under ongoing belimumab therapy.
Collapse
Affiliation(s)
- M Jüptner
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Klinik für Innere Medizin I, Kiel, Germany
| | | | | | | | | |
Collapse
|
23
|
Wang JJ, Chen C, Xie PF, Pan Y, Tan YH, Tang LJ. Proteomic analysis and immune properties of exosomes released by macrophages infected with Mycobacterium avium. Microbes Infect 2013; 16:283-91. [PMID: 24355714 DOI: 10.1016/j.micinf.2013.12.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 12/02/2013] [Accepted: 12/06/2013] [Indexed: 01/08/2023]
Abstract
The role of exosomes shed from Mycobacterium avium sp. paratuberculosis-infected macrophages in intercellular communication processes was examined. We compared the responses of resting macrophages infected with M. avium sp. paratuberculosis with those of resting macrophages treated with exosomes previously released from macrophages infected with M. avium sp. paratuberculosis. Some proteins components of exosomes released from resting macrophages infected with M. avium sp. paratuberculosis showed a significantly differential expression compared with exosomes from uninfected-macrophages. Both M. avium sp. paratuberculosis and exosomes from infected-cells enhanced the expression of CD80 and CD86 and the secretion of TNF-α and IFN-γ by macrophages. This suggests that exosomes from infected macrophages may be carriers of molecules, e.g. bacterial antigens and/or components from infected macrophages, that can elicit responses in resting cells. Two-dimensional analysis of the proteins present in exosomes from M. avium sp. paratuberculosis-infected macrophages compared with those from resting cells resulted in the identification by MALDI-TOF/TOF mass spectrometry of the following differentially expressed proteins: two actin isoforms, guanine nucleotide-binding protein β-1, cofilin-1 and peptidyl-prolyl cis-trans isomerase A. The possible relevance of the changes observed and the biological functions of the proteins differentially present are discussed.
Collapse
Affiliation(s)
- Jian-jun Wang
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha 410078, People's Republic of China
| | - Cai Chen
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha 410078, People's Republic of China
| | - Ping-fang Xie
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha 410078, People's Republic of China
| | - Yi Pan
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha 410078, People's Republic of China
| | - Yun-hong Tan
- Control Center of Tuberculosis in Hunan Province, Changsha 410205, People's Republic of China
| | - Li-jun Tang
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha 410078, People's Republic of China.
| |
Collapse
|
24
|
Treatment of SLE: Current paradigm and future directions. Clin Immunol 2013. [DOI: 10.1016/j.clim.2013.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|