1
|
New insights into IVIg mechanisms and alternatives in autoimmune and inflammatory diseases. Curr Opin Hematol 2021; 27:392-398. [PMID: 32868670 DOI: 10.1097/moh.0000000000000609] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Intravenous immunoglobulin (IVIg) is an effective treatment for an increasing number of autoimmune and inflammatory conditions. However, IVIg continues to be limited by problems of potential shortages and cost. A number of mechanisms have been described for IVIg, which have been captured in newly emergent IVIg mimetic and IVIg alternative therapies. This review discusses the recent developments in IVIg mimetics and alternatives. RECENT FINDINGS Newly emergent IVIg mimetics and alternatives capture major proposed mechanisms of IVIg, including FcγR blockade, FcRn inhibition, complement inhibition, immune complex mimetics and sialylated IgG. Many of these emergent therapies have promising preclinical and clinical trial results. SUMMARY Significant research has been undertaken into the mechanism of IVIg in the treatment of autoimmune and inflammatory disease. Understanding the major IVIg mechanisms has allowed for rational development of IVIg mimetics and alternatives for several IVIg-treatable diseases.
Collapse
|
2
|
Rotolo RA, Demuro J, Drummond G, Little C, Johns LD, Betz AJ. Prophylactic exposure to oral riluzole reduces the clinical severity and immune-related biomarkers of experimental autoimmune encephalomyelitis. J Neuroimmunol 2021; 356:577603. [PMID: 33992861 DOI: 10.1016/j.jneuroim.2021.577603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/26/2021] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Abstract
Glutamate-mediated excitotoxicity and immune cell infiltration are hallmarks of multiple sclerosis. The glutamate release inhibitor, riluzole (RIL), has been shown to attenuate the clinical symptoms of experimental autoimmune encephalomyelitis (EAE) in mice, but an association between glutamate excitotoxicity and the progression of MOG35-55-induced EAE has not been well defined. This study investigated the effects of prophylactic and chronic oral RIL on the clinical severity of EAE. Prophylactic+chronic RIL reduced the presence of inflammatory infiltrates, altered GFAP and Foxp3, and attenuated disease severity. These findings indicate a need to delineate the distinct role of glutamate in EAE symptomatology.
Collapse
Affiliation(s)
- Renee A Rotolo
- Department of Psychology, Quinnipiac University, Hamden, CT, United States of America; Biomedical Sciences, Quinnipiac University, Hamden, CT, United States of America; Department of Psychological Sciences, University of Connecticut, Storrs, CT, United States of America
| | - Jennifer Demuro
- Molecular and Cell Biology, Quinnipiac University, Hamden, CT, United States of America
| | - Gregory Drummond
- Biomedical Sciences, Quinnipiac University, Hamden, CT, United States of America
| | - Casey Little
- Department of Psychology, Quinnipiac University, Hamden, CT, United States of America
| | - Lennart D Johns
- Athletic Training, Quinnipiac University, Hamden, CT., United States of America; Lewis School of Health Sciences, Clarkson University, Potsdam, NY, United States of America
| | - Adrienne J Betz
- Department of Psychology, Quinnipiac University, Hamden, CT, United States of America.
| |
Collapse
|
3
|
Molina MF, Szyld D, Wilcox SR, Wittels KA. An Ominous Rash. J Emerg Med 2020; 59:435-438. [PMID: 32800638 DOI: 10.1016/j.jemermed.2020.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/01/2020] [Indexed: 06/11/2023]
Affiliation(s)
- Melanie F Molina
- Department of Emergency Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Demian Szyld
- Department of Emergency Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Department of Emergency Medicine, Harvard Medical School, Boston, Massachusetts
| | - Susan R Wilcox
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Emergency Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kathleen A Wittels
- Department of Emergency Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Department of Emergency Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
4
|
Achiron A, Miron S. Immunoglobulins Treatment in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. Mult Scler 2019. [DOI: 10.1177/135245850000602s02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Intravenously administered immunoglobulins (IgG) treatment has several modes of action that can regulate the immune response during different steps of the inflammatory process in experimental autoimmune encephalomyelitis (EAE) and Multiple Sclerosis (MS). The immunomodulatory effects IgG are largely dependent on their ability to interact with membrane molecules of lymphocytes and monocytes. Better understanding of these mechanisms of action in relation to the pathogenesis of MS, is important in order to decide the time of initiation and the duration of treatment in MS patients. In order to have the best beneficial effect on disease course, future research should focus on the initial events that activate the disease and on the early treatment modalities of IgG in MS.
Collapse
Affiliation(s)
- Anat Achiron
- Neuroimmunology MS Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Shmuel Miron
- Neuroimmunology MS Center, Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
5
|
Bernhardsson M, Dietrich-Zagonel F, Tätting L, Eliasson P, Aspenberg P. Depletion of cytotoxic (CD8+) T cells impairs implant fixation in rat cancellous bone. J Orthop Res 2019; 37:805-811. [PMID: 30737834 DOI: 10.1002/jor.24246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 01/29/2019] [Indexed: 02/04/2023]
Abstract
As cytotoxic (CD8+ ) T cells seem to impair shaft fracture healing, we hypothesized that depletion of CD8+ cells would instead improve healing of cancellous bone. Additionally, we also tested if CD8-depletion would influence the healing of ruptured Achilles tendons. Rats received a single injection of either anti-CD8 antibodies or saline and put through surgery 24 h later. Three different surgical interventions were performed as follows: (1) a drill hole in the proximal tibia with microCT (BV/TV) to assess bone formation; (2) a screw in the proximal tibia with mechanical evaluation (pull-out force) to assess fracture healing; (3) Achilles tendon transection with mechanical evaluation (force-at-failure) to assess tendon healing. Furthermore, CD8-depletion was confirmed with flow cytometry on peripheral blood. Flow cytometric analysis confirmed depletion of CD8+ cells (p < 0.001). Contrary to our hypothesis, depletion of CD8+ cells reduced the implant pull-out force by 19% (p < 0.05) and stiffness by 34% (p < 0.01), although the bone formation in the drill holes was the same as in the controls. Tendon healing was unaffected by CD8-depletion. Our results suggest that CD8+ cells have an important part in cancellous bone healing. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Magnus Bernhardsson
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Franciele Dietrich-Zagonel
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Love Tätting
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Pernilla Eliasson
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Per Aspenberg
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
6
|
Mouthon L, Kaveri SV, Spalter SH, Lacroix-Desmazes S, Lefranc C, Desai R, Kazatchkine MD. Mechanisms of action of intravenous immune globulin in immune-mediated diseases. Clin Exp Immunol 2019. [DOI: 10.1111/cei.1996.104.s1.3] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
7
|
Gelfand EW, Landwehr LP, Esterl B, Mazer B. Intravenous immune globulin: an alternative therapy in steroid-dependent allergic diseases. Clin Exp Immunol 2019. [DOI: 10.1111/cei.1996.104.s1.61] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
8
|
Achiron A, Barak Y, Goren M, Gabbay U, Miron S, Rotstein Z, Noy S, Sarova-Pinhas I. Intravenous immune globulin in multiple sclerosis: clinical and neuroradiological results and implications for possible mechanisms of action. Clin Exp Immunol 2019. [DOI: 10.1111/cei.1996.104.s1.67] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
9
|
Larose-Pierre M, Scrivens JJ, Norwood D, Rappa L. Necrotizing Fasciitis Caused by Group A Streptococcus: Case Report and Therapy Update. J Pharm Pract 2016. [DOI: 10.1106/bwr9-m77y-pqcv] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Necrotizing fasciitis is a life-threatening infection that affects the fascia and fat tissue underlying the skin. The diagnosis is often difficult because subcutaneous changes may not be readily apparent. Toxin-producing bacteria are usually the cause, with group A streptococcus (GAS) or Streptococcus pyogenes being responsible for a significant portion of the morbidity and mortality associated with this infection. The mortality rate associated with necrotizing fasciitis varies between 30% and 60%. Toxic shock-like syndrome and multisystem organ failure are the usual causes of death. Early diagnosis and surgery have been associated with decreased morbidity and mortality, and appropriate antimicrobial (eg, penicillin plus clindamycin) and supportive therapy is of utmost importance. Intravenous immunoglobulin and hyperbaric oxygen therapy may be beneficial in treating the infection; however, these 2 therapies require further research. Clinicians need to familiarize themselves with the disease and the different treatment modalities to be able to make the appropriate therapeutic decision. The optimal treatment of necrotizing fasciitis still remains a challenge today. This article presents an illustrative case with a brief overview of necrotizing fasciitis, and the current therapeutic modalities used in the management of the disease.
Collapse
Affiliation(s)
- Margareth Larose-Pierre
- Florida A &M University, College of Pharmacy and Pharmaceutical Sciences, Miami Division; and Clinical Pharmacy Specialist, Surgical Intensive Care, Department of Pharmacy, Veterans Affairs Medical Center, Miami, Florida,
| | - John J. Scrivens
- Florida A &M University, College of Pharmacy and Pharmaceutical Sciences, Tampa Bay Division, Tampa, Florida,
| | - Daryl Norwood
- Pharmacy Practice, Florida A &M University, College of Pharmacy and Pharmaceutical Sciences, Miami Division, Miami, Florida,
| | - Leonard Rappa
- Pharmacy Practice, Florida A & M University, College of Pharmacy and Pharmaceutical Sciences, Miami Division, Miami, Florida,
| |
Collapse
|
10
|
Petereit HF, Reske D, Pukrop R, Maas-Enriquez M, Japp G, Jongen PJH, Kölmel HW, Merkelbach S, Hartung HP, Heiss WD, Hommes OR. No effect of intravenous immunoglobulins on cytokine-producing lymphocytes in secondary progressive multiple sclerosis. Mult Scler 2016; 12:66-71. [PMID: 16459721 DOI: 10.1191/135248506ms1246oa] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Intravenous immunoglobulins (IVIG) have been effective in reducing multiple sclerosis (MS) disease activity and improving disability scores. However, the mechanism by which this beneficial effect is achieved remains unclear. An effect of IVIG on pro- and anti-inflammatory cytokines- which are thought to play a role in the disease process- has been postulated in a number of animal and ex vivo studies. Hence, we performed a study on 34 patients with secondary progressive (SP) MS being treated with monthly IVIG or placebo for two years according to the protocol of the ESIMS study. Clinical outcome measures and cytokine production (interferon gamma, tumour necrosis factor alpha, interleukin-4 and -10) were recorded in all patients and compared with respect to the treatment group. Against our expectations, IVIG did not reduce the relapse rate or the progression of disability or cytokine production. Our data argue against an enduring immunomodulating effect of IVIG, at least in SPMS.
Collapse
Affiliation(s)
- H F Petereit
- Department of Neurology and Psychiatry, University of Cologne, Josef-Stelzmann-Str. 9, 50924 Kö1n, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Quast I, Keller CW, Weber P, Schneider C, von Gunten S, Lünemann JD. Protection from experimental autoimmune encephalomyelitis by polyclonal IgG requires adjuvant-induced inflammation. J Neuroinflammation 2016; 13:42. [PMID: 26893156 PMCID: PMC4758141 DOI: 10.1186/s12974-016-0506-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/08/2016] [Indexed: 11/11/2022] Open
Abstract
Background Intravenous immunoglobulin (IVIG) proved to be an efficient anti-inflammatory treatment for a growing number of neuroinflammatory diseases and protects against the development of experimental autoimmune encephalomyelitis (EAE), a widely used animal model for multiple sclerosis (MS). Methods The clinical efficacy of IVIG and IVIG-derived F(ab’)2 fragments, generated using the streptococcal cysteine proteinase Ide-S, was evaluated in EAE induced by active immunization and by adoptive transfer of myelin-specific T cells. Frequency, phenotype, and functional characteristics of T cell subsets and myeloid cells were determined by flow cytometry. Antibody binding to microbial antigen and cytokine production by innate immune cells was assessed by ELISA. Results We report that the protective effect of IVIG is lost in the adoptive transfer model of EAE and requires prophylactic administration during disease induction. IVIG-derived Fc fragments are not required for protection against EAE, since administration of F(ab’)2 fragments fully recapitulated the clinical efficacy of IVIG. F(ab’)2-treated mice showed a substantial decrease in splenic effector T cell expansion and cytokine production (GM-CSF, IFN-γ, IL-17A) 9 days after immunization. Inhibition of effector T cell responses was not associated with an increase in total numbers of Tregs but with decreased activation of innate myeloid cells such as neutrophils, monocytes, and dendritic cells. Therapeutically effective IVIG-derived F(ab’)2 fragments inhibited adjuvant-induced innate immune cell activation as determined by IL-12/23 p40 production and recognized mycobacterial antigens contained in Freund’s complete adjuvant which is required for induction of active EAE. Conclusions Our data indicate that F(ab’)2-mediated neutralization of adjuvant contributes to the therapeutic efficacy of anti-inflammatory IgG. These findings might partly explain the discrepancy of IVIG efficacy in EAE and MS.
Collapse
Affiliation(s)
- Isaak Quast
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| | - Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| | - Patrick Weber
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| | | | | | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
12
|
Zaaqoq AM, Khasawneh FA, Smalligan RD. Cardiovascular Complications of HIV-Associated Immune Dysfunction. Cardiol Res Pract 2015; 2015:302638. [PMID: 25648075 PMCID: PMC4305617 DOI: 10.1155/2015/302638] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/27/2014] [Indexed: 12/25/2022] Open
Abstract
Prolonged survival in HIV infection is accompanied by an increased frequency of non-HIV-related comorbidities. It is suggested that cardiovascular diseases (CVD) occur earlier among HIV-positive patients compared with HIV-negative patients, and at a higher rate. Several factors have been proposed which can be categorized into traditional and nontraditional risk factors. Immune dysfunction is a nontraditional risk factor that contributes significantly to cardiovascular pathology. Markers of inflammation are elevated in HIV-infected patients, and elevations in markers such as high-sensitivity C-reactive protein, D-dimer, and interleukin-6 (IL-6) have been associated with increased risk for cardiovascular disease. However, the data currently suggest the most practical advice is to start antiretroviral therapy early and to manage traditional risk factors for CVD aggressively. A better understanding of the mechanisms of CVD in this population and further efforts to modify chronic inflammation remain an important research area.
Collapse
Affiliation(s)
- Akram M. Zaaqoq
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Faisal A. Khasawneh
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Division of Infectious Diseases, Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Roger D. Smalligan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
13
|
High-dose IgG therapy mitigates bile duct-targeted inflammation and obstruction in a mouse model of biliary atresia. Pediatr Res 2014; 76:72-80. [PMID: 24727948 PMCID: PMC4062601 DOI: 10.1038/pr.2014.46] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/27/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND A proposed etiology of biliary atresia (BA) entails a virus-induced, progressive immune-mediated injury of the biliary system. Intravenous Ig (IVIg) has demonstrated clinical benefit in several inflammatory diseases. The aim of this study was to determine the therapeutic effects of high-dose IgG treatment in the rhesus rotavirus (RRV)-induced mouse model of BA. METHODS Newborn mice were infected with RRV, and jaundiced mice were given high-dose IgG or albumin control. Survival, histology, direct bilirubin, liver immune cell subsets, and cytokine production were analyzed. RESULTS There was no difference in overall survival between RRV-infected groups, however high-dose IgG resulted in decreased bilirubin, bile duct inflammation, and increased extrahepatic bile duct patency. High-dose IgG decreased vascular cell adhesion molecule-1, resulting in limited migration of immune cells to portal tracts. High-dose IgG significantly decreased CD4(+) T cell production of interleukin (IL)-2, interferon (IFN)-γ, and tumor necrosis factor (TNF)-α and CD8(+) T cell production of IFN-γ, as well as increased levels of regulatory T cells. CONCLUSION High-dose IgG therapy in murine BA dramatically decreased Th1 cell-mediated inflammation and biliary obstruction. This study lends support for consideration of IVIg clinical trials in infants with BA, to diminish the progressive intrahepatic bile duct injury.
Collapse
|
14
|
Campbell IK, Miescher S, Branch DR, Mott PJ, Lazarus AH, Han D, Maraskovsky E, Zuercher AW, Neschadim A, Leontyev D, McKenzie BS, Käsermann F. Therapeutic effect of IVIG on inflammatory arthritis in mice is dependent on the Fc portion and independent of sialylation or basophils. THE JOURNAL OF IMMUNOLOGY 2014; 192:5031-8. [PMID: 24760152 DOI: 10.4049/jimmunol.1301611] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
High-dose i.v. Ig (IVIG) is used to treat various autoimmune and inflammatory diseases; however, the mechanism of action remains unclear. Based on the K/BxN serum transfer arthritis model in mice, IVIG suppression of inflammation has been attributed to a mechanism involving basophils and the binding of highly sialylated IgG Fc to DC-SIGN-expressing myeloid cells. The requirement for sialylation was examined in the collagen Ab-induced arthritis (CAbIA) and K/BxN serum transfer arthritis models in mice. High-dose IVIG (1-2 g/kg body weight) suppressed inflammatory arthritis when given prophylactically. The same doses were also effective in the CAbIA model when given subsequent to disease induction. In this therapeutic CAbIA model, the anti-inflammatory effect of IVIG was dependent on IgG Fc but not F(ab')2 fragments. Removal of sialic acid residues by neuraminidase had no impact on the anti-inflammatory activity of IVIG or Fc fragments. Treatment of mice with basophil-depleting mAbs did not abrogate the suppression of either CAbIA or K/BxN arthritis by IVIG. Our data confirm the therapeutic benefit of IVIG and IgG Fc in Ab-induced arthritis but fail to support the significance of sialylation and basophil involvement in the mechanism of action of IVIG therapy.
Collapse
Affiliation(s)
- Ian K Campbell
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Sylvia Miescher
- CSL Behring, Research and Development, CH-3000 Bern, Switzerland
| | - Donald R Branch
- Canadian Blood Services, Centre for Innovation, Toronto, Ontario K1G 4J5, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5G 2M1, Canada; and
| | - Patrick J Mott
- Canadian Blood Services, Centre for Innovation, Toronto, Ontario K1G 4J5, Canada; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Alan H Lazarus
- Canadian Blood Services, Centre for Innovation, Toronto, Ontario K1G 4J5, Canada; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Dongji Han
- Canadian Blood Services, Centre for Innovation, Toronto, Ontario K1G 4J5, Canada; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | | | - Adrian W Zuercher
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia; CSL Behring, Research and Development, CH-3000 Bern, Switzerland
| | - Anton Neschadim
- Canadian Blood Services, Centre for Innovation, Toronto, Ontario K1G 4J5, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5G 2M1, Canada; and
| | - Danila Leontyev
- Canadian Blood Services, Centre for Innovation, Toronto, Ontario K1G 4J5, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5G 2M1, Canada; and
| | | | - Fabian Käsermann
- CSL Behring, Research and Development, CH-3000 Bern, Switzerland;
| |
Collapse
|
15
|
Saito E, Koike T, Hashimoto H, Miyasaka N, Ikeda Y, Hara M, Yamada H, Yoshida T, Harigai M, Ichikawa Y. Efficacy of high-dose intravenous immunoglobulin therapy in Japanese patients with steroid-resistant polymyositis and dermatomyositis. Mod Rheumatol 2014. [DOI: 10.3109/s10165-007-0013-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
16
|
Essential role of NK cells in IgG therapy for experimental autoimmune encephalomyelitis. PLoS One 2013; 8:e60862. [PMID: 23577171 PMCID: PMC3618232 DOI: 10.1371/journal.pone.0060862] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 03/04/2013] [Indexed: 12/25/2022] Open
Abstract
Intravenous immunoglobulin has long been used in treating autoimmune diseases, although mechanisms remain uncertain. Activating Fcγ receptors are receptors of IgG and reported to be essential in intravenous immunoglobulin (IVIG) therapy. Therefore, we hypothesized natural killer (NK) cells, which express abundant activating Fcγ receptors, are the potential cellular target. In experimental autoimmune encephalomyelitis (EAE), we demonstrated that IgG suppressed disease development in intact, but not in NK cell depleted mice. Adoptive transfer of IgG-treated NK cell could protect mice against EAE, and suppressed interferon γ and interleukin 17 production. The percentage of CD4+Foxp3+ regulatory T cells was significantly increased. The increase of regulatory T cells was also observed in IgG-treated EAE mice but not in NK cell depleted mice. In vitro experiments confirmed that IgG-treated NK cells enhanced regulatory T cell induction from naïve CD4+ T cells. Interestingly, cells from draining lymph nodes produced more interleukin 2 after the adoptive transfer of IgG-treated NK cells. We neutralized interleukin 2 and the induction of CD4+Foxp3+ T cells by IgG-treated NK cells was significantly reduced. To our knowledge, we identified for the first time the critical role of NK cells in the mechanism of IgG-induced induction of Treg cells in treatment of autoimmunity.
Collapse
|
17
|
Othy S, Hegde P, Topçu S, Sharma M, Maddur MS, Lacroix-Desmazes S, Bayry J, Kaveri SV. Intravenous Gammaglobulin Inhibits Encephalitogenic Potential of Pathogenic T Cells and Interferes with their Trafficking to the Central Nervous System, Implicating Sphingosine-1 Phosphate Receptor 1–Mammalian Target of Rapamycin Axis. THE JOURNAL OF IMMUNOLOGY 2013; 190:4535-41. [DOI: 10.4049/jimmunol.1201965] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
18
|
Padet L, Bazin R. IVIg prevents the in vitro activation of T cells by neutralizing the T cell activators. Immunol Lett 2013; 150:54-60. [PMID: 23295311 DOI: 10.1016/j.imlet.2012.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 12/14/2012] [Accepted: 12/27/2012] [Indexed: 11/26/2022]
Abstract
Clinical observations in patients treated with IVIg revealed significant modulations in T cell populations and functions. However, it is unclear whether IVIg acts directly on activated T cells to suppress their functions. To clarify the exact mechanism of IVIg action, we studied its effect on T cells activated using anti-CD3/CD28 microbeads to mimic stimulatory signals provided by accessory cells. We report here that IVIg reduces T cell proliferation and cytokine secretion by interfering with the ability of anti-CD3/CD28 microbeads to deliver activating signals to T cells. We further show that the interference occurs between IVIg and anti-CD3/CD28 microbeads and does not involve T cells. In conclusion, our work suggests that T cells are not a direct target of IVIg and that the modulation of T cell populations and functions observed in treated patients is the indirect consequence of a direct effect of IVIg on accessory cells.
Collapse
Affiliation(s)
- Lauriane Padet
- Department of Research and Development, Héma-Québec, Québec, Canada.
| | | |
Collapse
|
19
|
Okuda S, Kamei S, Harano S, Shinya N, Hayashida K, Sasaki T. [Enhancement of regulatory T cell induction by intravenous S-sulfonated Immunoglobulin during the treatment of experimental autoimmune encephalomyelitis]. YAKUGAKU ZASSHI 2012; 132:243-9. [PMID: 22293707 DOI: 10.1248/yakushi.132.243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intravenous immunoglobulin (IVIg) has been shown to be effective for a variety of autoimmune diseases. Despite its widespread use and therapeutic success, the precise mechanisms for the anti-inflammatory therapeutic effects of IVIg are not well understood. In particular, few reports have examined the mechanism of IVIg on regulatory T cells (Treg: CD4(+)CD25(+)FoxP3(+) T cells). In the present study, to clarify the effect of intravenous S-sulfonated immunoglobulin (S-IVIg) on Treg, we investigated experimental autoimmune encephalomyelitis (EAE), the representative animal model of autoimmune disease. First, when we evaluated the effect of S-IVIg in an acute EAE model, the prophylactic treatment of S-IVIg dose-dependently controlled the symptoms of EAE. Next, we measured Treg in EAE mice spleen by flow cytometry. The percentage of Treg in S-IVIg-treated mice was significantly increased compared with Saline-treated mice. Finally, in reinduced EAE, S-IVIg not only prevented EAE progression, but also increased the percentage of Treg in the spleen. The increase in percentage of Treg in S-IVIg-treated EAE might be associated with protection against EAE. These observations provide important evidence that IVIg is effective in T-cell-mediated control of autoimmunity.
Collapse
Affiliation(s)
- Sachio Okuda
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN), Kumamoto, Japan.
| | | | | | | | | | | |
Collapse
|
20
|
Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2012; 164:1079-106. [PMID: 21371012 DOI: 10.1111/j.1476-5381.2011.01302.x] [Citation(s) in RCA: 1014] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple sclerosis (MS). EAE is a complex condition in which the interaction between a variety of immunopathological and neuropathological mechanisms leads to an approximation of the key pathological features of MS: inflammation, demyelination, axonal loss and gliosis. The counter-regulatory mechanisms of resolution of inflammation and remyelination also occur in EAE, which, therefore can also serve as a model for these processes. Moreover, EAE is often used as a model of cell-mediated organ-specific autoimmune conditions in general. EAE has a complex neuropharmacology, and many of the drugs that are in current or imminent use in MS have been developed, tested or validated on the basis of EAE studies. There is great heterogeneity in the susceptibility to the induction, the method of induction and the response to various immunological or neuropharmacological interventions, many of which are reviewed here. This makes EAE a very versatile system to use in translational neuro- and immunopharmacology, but the model needs to be tailored to the scientific question being asked. While creating difficulties and underscoring the inherent weaknesses of this model of MS in straightforward translation from EAE to the human disease, this variability also creates an opportunity to explore multiple facets of the immune and neural mechanisms of immune-mediated neuroinflammation and demyelination as well as intrinsic protective mechanisms. This allows the eventual development and preclinical testing of a wide range of potential therapeutic interventions.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neurology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | | | | | | |
Collapse
|
21
|
Padet L, St-Amour I, Aubin É, Bazin R. Neutralization of mitogenic lectins by intravenous immunoglobulin (IVIg) prevents T cell activation: does IVIg really have a direct effect on T cells? Clin Exp Immunol 2012; 166:352-60. [PMID: 22059993 DOI: 10.1111/j.1365-2249.2011.04476.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Intravenous immunoglobulin (IVIg) is used for the treatment of an increasing number of autoimmune diseases. Clinical observations on IVIg-treated patients have revealed a modulation of T cell populations and functions in these patients. In vitro studies aimed at understanding the mechanisms underlying the effects of IVIg on T cells led to the conclusion that IVIg directly affected lectin-activated T cell functions. However, more recent studies have suggested the absence of a direct effect of IVIg on T cells. In the present work, we revisited the effect of IVIg on T cells using lectin-stimulated human T cells and showed that IVIg inhibited T cell functions only when added simultaneously with the activating lectin. Further, we showed that IVIg depleted from lectin-reactive IgG was no longer inhibitory, suggesting that the effect of IVIg on T cells was the consequence of lectin neutralization, possibly by interaction with glycans present in F(ab')(2) portion of IgG molecules. Our results challenge the previously widely accepted notion that IVIg exerts its anti-inflammatory effects by acting directly on T cells and suggest that effects of IVIg observed in treated patients are rather a consequence of the recently reported inhibitory effect of IVIg on antigen presentation.
Collapse
Affiliation(s)
- L Padet
- Department of Research and Development, Héma-Québec, Québec, QC, Canada
| | | | | | | |
Collapse
|
22
|
Multi-faceted role of naturally occurring autoantibodies in fighting pathogens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 750:100-13. [PMID: 22903669 DOI: 10.1007/978-1-4614-3461-0_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Naturally occurring antibodies (NAbs) play a vital role in the first line of defense against bacterial and viral infections. Most studies in mice and man have attributed this role to NAbs of the IgM isotype. However, there is also a significant amount of data on the anti-infectious function of NAbs of the IgG isotype. Most of these observations are derived from studies using a privileged source of NAbs, the pooled human IgG for intravenous application, IVIG. In addition to its use as a replacement in humoral immunodeficiencies, IVIG is extensively used in autoimmune and inflammatory diseases. The properties of NAbs, the principal components of IVIG, are considered crucial for their immune-regulatory properties, owing to their ability to recognize self-antigens and even autoantibodies. By virtue of these specificities for several cellular antigens, including exposed proteins that act as receptors for a variety of pathogens, certain NAbs in IVIG have a therapeutic role in preventing or modulating infections. We summarize in this chapter several examples that highlight the importance of NAbs in the control of certain bacterial and viral infections.
Collapse
|
23
|
Gregoire-Gauthier J, Durrieu L, Duval A, Fontaine F, Dieng MM, Bourgey M, Patey-Mariaud de Serre N, Louis I, Haddad E. Use of immunoglobulins in the prevention of GvHD in a xenogeneic NOD/SCID/γc- mouse model. Bone Marrow Transplant 2011; 47:439-50. [PMID: 21572464 DOI: 10.1038/bmt.2011.93] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The efficacy of IVIG in preventing GvHD has not been definitely demonstrated clinically. Using a xenogeneic model of GvHD in NOD/SCID/γc- (NSG) mice, we showed that weekly administration of IVIG significantly reduced the incidence and associated mortality of GvHD to a degree similar to CsA. Unlike CsA and OKT3, IVIG were not associated with inhibition of human T-cell proliferation in mice. Instead, IVIG significantly inhibited the secretion of human IL-17, IL-2, IFN-γ and IL-15 suggesting that IVIG prevented GvHD by immunomodulation. Furthermore, the pattern of modification of the human cytokine storm differed from that observed with CsA and OKT3. Finally, in a humanized mouse model of immune reconstitution, in which NSG mice were engrafted with human CD34(+) stem cells, IVIG transiently inhibited B-cell reconstitution, whereas peripheral T-cell reconstitution and thymopoiesis were unaffected. Together these in vivo data raise debate related to the appropriateness of IVIG in GvHD prophylaxis. In addition, this model provides an opportunity to further elucidate the precise mechanism(s) by which IVIG inhibit GvHD.
Collapse
Affiliation(s)
- J Gregoire-Gauthier
- CHU Sainte-Justine Research Center, Center de Cancérologie Charles-Bruneau, 3175 chemin de la Côte-Ste-Catherine, Montréal, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Pavlovic S, Zdravkovic N, Dimitrov JD, Djukic A, Arsenijevic N, Vassilev TL, Lukic ML. Intravenous immunoglobulins exposed to heme (heme IVIG) are more efficient than IVIG in attenuating autoimmune diabetes. Clin Immunol 2011; 138:162-71. [DOI: 10.1016/j.clim.2010.10.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 10/07/2010] [Accepted: 10/29/2010] [Indexed: 01/11/2023]
|
25
|
|
26
|
Indirect inhibition of in vivo and in vitro T-cell responses by intravenous immunoglobulins due to impaired antigen presentation. Blood 2009; 115:1727-34. [PMID: 19965673 DOI: 10.1182/blood-2009-06-225417] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several clinical studies done with intravenous immunoglobulin (IVIg)-treated autoimmune patients as well as several in vitro studies have revealed that IVIg can reduce polyclonal T-cell activation and modify their cytokine secretion pattern. However, their effect on (auto)antigen-specific T-cell responses has never been addressed directly. In the present work, we used an in vivo model of induction of antigen-specific T-cell responses and an in vitro antigen presentation system to study the effects of IVIg on T-cell responses. The results obtained showed that IVIg inhibited both the in vivo and in vitro antigen-specific T-cell responses but that this effect was the indirect consequence of a reduction in the antigen presentation ability of antigen-presenting cells. The inhibitory effect of IVIg was FcgammaRIIb-independent, suggesting that IVIg must interfere with activating FcgammaRs expressed on antigen-presenting cells to reduce their ability to present antigens. Such inhibition of T-cell responses by reducing antigen presentation may therefore contribute to the well-known anti-inflammatory effects of IVIg in autoimmune diseases.
Collapse
|
27
|
Graber JJ, Dhib-Jalbut S. Protective autoimmunity in the nervous system. Pharmacol Ther 2008; 121:147-59. [PMID: 19000712 DOI: 10.1016/j.pharmthera.2008.10.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 10/02/2008] [Indexed: 12/31/2022]
Abstract
The immune system can play both detrimental and beneficial roles in the nervous system. Multiple arms of the immune system, including T cells, B cells, NK cells, mast cells, macrophages, dendritic cells, microglia, antibodies, complement and cytokines participate in limiting damage to the nervous system during toxic, ischemic, hemorrhagic, infective, degenerative, metabolic and immune-mediated insults and also assist in the process of repair after injury has occurred. Immune cells have been shown to produce neurotrophic growth factors and interact with neurons and glial cells to preserve them from injury and stimulate growth and repair. The immune system also appears to participate in proliferation of neural progenitor stem cells and their migration to sites of injury. Neural stem cells can also modify the immune response in the central and peripheral nervous system to enhance neuroprotective effects. Evidence for protective and reparative functions of the immune system has been found in diverse neurologic diseases including traumatic injury, ischemic and hemorrhagic stroke, multiple sclerosis, infection, and neurodegenerative diseases (Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis). Existing therapies including glatiramer acetate, interferon-beta and immunoglobulin have been shown to augment the protective and regenerative aspects of the immune system in humans, and other experimental interventions such as vaccination, minocycline, antibodies and neural stem cells, have shown promise in animal models of disease. The beneficent aspects of the immune response in the nervous system are beginning to be appreciated and their potential as pharmacologic targets in neurologic disease is being explored.
Collapse
Affiliation(s)
- Jerome J Graber
- New York University School of Medicine, Department of Neurology, New York, NY, USA
| | | |
Collapse
|
28
|
Makay B, Yilmaz S, Türkyilmaz Z, Unal N, Oren H, Unsal E. Etanercept for therapy-resistant macrophage activation syndrome. Pediatr Blood Cancer 2008; 50:419-21. [PMID: 16900487 DOI: 10.1002/pbc.21019] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Macrophage activation syndrome (MAS) is a severe, potentially fatal complication of childhood rheumatic diseases, especially systemic onset juvenile idiopathic arthritis (SoJIA). We report a 4-year-old girl with probable SoJIA who presented with MAS. She did not respond to pulse methyl prednisolone and Cyclosporine A (CsA). She also failed to respond to intravenous immunoglobulin (IVIG) therapy. Etanercept was started, based on the observation of increased serum levels of tumor necrosis factor-alpha (TNF-alpha) in patients with MAS. Her condition improved following etanercept, suggesting that etanercept might have a therapeutic role in resistant MAS.
Collapse
Affiliation(s)
- Balahan Makay
- Department of Pediatric Rheumatology, Dokuz Eylul University Faculty of Medicine, Balcova, Izmir, Turkey.
| | | | | | | | | | | |
Collapse
|
29
|
Efficacy of high-dose intravenous immunoglobulin therapy in Japanese patients with steroid-resistant polymyositis and dermatomyositis. Mod Rheumatol 2008; 18:34-44. [PMID: 18217197 DOI: 10.1007/s10165-007-0013-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 09/26/2007] [Indexed: 10/22/2022]
Abstract
Intravenous immunoglobulin (IVIG) therapy was administered to 15 patients who were refractory to traditional steroid therapy [eight with polymyosis (PM), seven with dermamyosis (DM)] to evaluate its efficacy. Serum creatine kinase (CK) significantly decreased from week 1, and manual muscle test scores (MMT) and activities of daily living (ADL) significantly increased from week 2. Efficacy rates were 93.3% (14/15 patients) as assessed using the MMT score, 80.0% (12/15 patients) using the ADL score, and 100% (15/15 patients) using the serum CK level. When changes in the serum CK level over two four-week periods, one before IVIG therapy (from week -4 to week 0) and one after IVIG therapy (from week 0 to week 4), were transformed to natural logarithms, the four-week change after IVIG therapy was significantly greater than that before IVIG therapy. The estimated duration of the serum CK level remaining normal in 50% of the patients after IVIG therapy was 334.5 days. Adverse reactions were observed in seven of 16 patients (43.8%) during the study period, but none of the adverse reactions were considered to be serious or required emergency treatment. In conclusion, the present study indicates that IVIG therapy is effective for steroid-resistant PM/DM.
Collapse
|
30
|
Expansion of CD4+CD25+ regulatory T cells by intravenous immunoglobulin: a critical factor in controlling experimental autoimmune encephalomyelitis. Blood 2008; 111:715-22. [DOI: 10.1182/blood-2007-03-079947] [Citation(s) in RCA: 227] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The clinical use of intravenous immunoglobulin (IVIg) based on its immunomodulatory and anti-inflammatory potential remains an ongoing challenge. Fcγ receptor-mediated effects of IVIg, although well elucidated in certain pathologies, cannot entirely account for its proven benefit in several autoimmune disorders mediated by autoreactive T cells. In this study, we show that prophylactic infusion of IVIg prevents the development of experimental autoimmune encephalomyelitis (EAE), an accepted animal model for multiple sclerosis (MS). The protection was associated with peripheral increase in CD4+CD25+Foxp3+ regulatory T cell (Treg) numbers and function. The protection was Treg-mediated because IVIg failed to protect against EAE in mice that were depleted of the Treg population. Rather than inducing de novo generation from conventional T cells, IVIg had a direct effect on proliferation of natural Treg. In conclusion, our results highlight a novel mechanism of action of IVIg and provide a rationale to test the use of IVIg as an immunomodulatory tool to enhance Treg in early onset MS and other autoimmune and inflammatory conditions.
Collapse
|
31
|
Clynes R. Protective mechanisms of IVIG. Curr Opin Immunol 2007; 19:646-51. [DOI: 10.1016/j.coi.2007.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 09/02/2007] [Indexed: 11/17/2022]
|
32
|
Fuchs S, Feferman T, Zhu KY, Meidler R, Margalit R, Wang N, Laub O, Souroujon MC. Suppression of Experimental Autoimmune Myasthenia Gravis by Intravenous Immunoglobulin and Isolation of a Disease-Specific IgG Fraction. Ann N Y Acad Sci 2007; 1110:550-8. [PMID: 17911471 DOI: 10.1196/annals.1423.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Intravenous immunoglobulin (IVIG) administration has been beneficially used for the treatment of a variety of autoimmune diseases including myasthenia gravis (MG). We have demonstrated that IVIG administration in experimental autoimmune MG (EAMG) results in suppression of disease that is accompanied by decreased Th1 cell and B cell proliferation. Chromatography of pooled human immunoglobulins (IVIGs) on immobilized IgG, isolated from rats with EAMG, results in a complete depletion of the suppressive activity of the IVIG. Moreover, the eluate from this EAMG-specific antibody column retains the immunosuppressive activity of IVIG. This study supports the notion that the therapeutic effect of IVIGs is mediated by an antigen-specific anti-immunoglobulin (anti-idiotypic) activity that is essential for its suppressive activity.
Collapse
Affiliation(s)
- Sara Fuchs
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Sticherling M, Trawinski H. Effects of Intravenous Immunoglobulins on Peripheral Blood Mononuclear Cell Activation in Vitro. Ann N Y Acad Sci 2007; 1110:694-708. [PMID: 17911484 DOI: 10.1196/annals.1423.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The therapeutic effects of intravenous immunoglobulins (IVIGs) on different chronic inflammatory and autoimmune diseases is well appreciated, though clinical studies with high-evidence levels are largely missing. Similar to the broad spectrum of diseases and their underlying etiopathogenic background, the mechanisms of action seem heterogenous and multifold. Several studies addressing in vitro and in vivo effects of IVIG on various immunological parameters have been described with partly contradictory results. In this study immunoglobulins and stabilizers present in commercial IVIG preparations were studied in regard to the in vitro proliferation and cytokine production of peripheral blood mononuclear cells when stimulated with phytohemagglutinin (PHA), interleukin 2, and tetanus toxoid. Whereas the immunoglobulins stimulate the proliferation of PBMCs and decrease IFNgamma secretion, stabilizers of IVIG seem to inhibit the proliferation of PBMCs while increasing the secretion of IFN gamma. These effects have to be taken into account when balancing the impact of IVIG dosage and infusion intervals and relating them to clinical side effects and therapeutic efficacy.
Collapse
Affiliation(s)
- Michael Sticherling
- Hautklinik, Universitätsklinikum Erlangen, Hartmannstr. 14, D-91952 Erlangen, Germany.
| | | |
Collapse
|
34
|
Tristano AG, Casanova-Escalona L, Torres A, Rodríguez MA. Macrophage activation syndrome in a patient with systemic onset rheumatoid arthritis: rescue with intravenous immunoglobulin therapy. J Clin Rheumatol 2007; 9:253-8. [PMID: 17041466 DOI: 10.1097/01.rhu.0000081259.61370.eb] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We report the case of a 7-year-old boy with systemic onset juvenile rheumatoid arthritis (JRA) who developed macrophage activation syndrome during a period of relative remission. He presented with high-grade fever, enlarged lymph nodes, acute hepatitis, severe pancytopenia, and evidence of disseminated intravascular coagulopathy. He became severely ill, with persistent mucosal bleeding, hypovolemia, and vascular instability, and he was admitted to the intensive care unit. No response was observed to treatment with high-dose intravenous corticosteroids and blood product replacements, but the patient fully recovered after the completion of two pulses of intravenous immunoglobulin. This treatment modality may be an alternative to anti-TNF-alpha and cyclosporine A for a prompt response in JRA patients with this potentially fatal condition.
Collapse
Affiliation(s)
- Antonio G Tristano
- Centro Nacional de Enfermedades Reumáticas, Hospital Universitario de Caracas, Caracas, Venezuela
| | | | | | | |
Collapse
|
35
|
Lin HH, Spies JM, Lu JL, Pollard JD. Effective treatment of experimental autoimmune neuritis with human immunoglobulin. J Neurol Sci 2007; 256:61-7. [PMID: 17379248 DOI: 10.1016/j.jns.2007.02.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 02/06/2007] [Accepted: 02/12/2007] [Indexed: 10/23/2022]
Abstract
High-dose intravenous immunoglobulin (IVIg) is an effective treatment for inflammatory demyelinating neuropathies, although the mechanism(s) of action remain incompletely understood. Experimental autoimmune neuritis (EAN) is an animal model of inflammatory demyelinating neuropathies; however, there have been conflicting reports regarding the efficacy of human IVIg in EAN. To obtain a model suitable for the study of the mechanism(s) of action of IVIg in Guillain-Barré syndrome, we investigated the effect of IVIg in EAN in the rat using clinical, electrophysiological and morphological measures. Human IVIg administered at the onset of signs of disease proved effective in preventing further progression of disease and shortening disease duration. This effectiveness was associated with significant differences in electrophysiological parameters including less prolongation of somatosensory evoked potential (S wave) latencies, better maintained S wave amplitudes, less reduction of distal motor nerve conduction velocity, and better maintained amplitudes of compound muscle action potentials of the dorsal foot muscles after stimulation at ankle and hip. Moreover, treatment with IVIg resulted in significantly lower histological grades in rat EAN. The current study provides evidence that human IVIg is effective in the treatment of EAN in the rat, indicating that this model may facilitate further investigation of the mechanism(s) of action of IVIg in inflammatory demyelinating neuropathies.
Collapse
MESH Headings
- Action Potentials/drug effects
- Action Potentials/physiology
- Animals
- Disease Models, Animal
- Electromyography/methods
- Evoked Potentials, Somatosensory/drug effects
- Evoked Potentials, Somatosensory/radiation effects
- Humans
- Immunoglobulins, Intravenous/therapeutic use
- Male
- Neural Conduction/drug effects
- Neural Conduction/physiology
- Neuritis, Autoimmune, Experimental/drug therapy
- Neuritis, Autoimmune, Experimental/pathology
- Neuritis, Autoimmune, Experimental/physiopathology
- Rats
- Rats, Inbred Lew
- Reaction Time/drug effects
- Tolonium Chloride
Collapse
Affiliation(s)
- Hsin Hsin Lin
- Department of Medicine, The University of Sydney, Sydney, Australia.
| | | | | | | |
Collapse
|
36
|
Seiwa C, Yamamoto M, Tanaka K, Fukutake M, Ueki T, Takeda S, Sakai R, Ishige A, Watanabe K, Akita M, Yagi T, Tanaka K, Asou H. Restoration of FcRγ/Fyn signaling repairs central nervous system demyelination. J Neurosci Res 2007; 85:954-66. [PMID: 17290413 DOI: 10.1002/jnr.21196] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Disruption of myelin causes severe neurological diseases. An understanding of the mechanisms that control myelination and remyelination is needed to develop therapeutic strategies for demyelinating diseases such as multiple sclerosis (MS). Our previous finding indicating the critical involvement of the gamma chain of immunogloblin Fc receptors (FcRgamma) and Fyn signaling in oligodendrocyte differentiaion and myelination demands a fundamental revision of the strategies used for MS therapy, because antigen-antibody complexes in MS patients may induce the direct dysregulation of myelination process as well as the inflammatory destruction of myelin sheath. Here we show that the FcRgamma/Fyn signaling cascade is critically involved in cuprizone-induced demyelination/remyelination, with no lymphocytic response. The levels of phosphorylated myelin basic proteins (p-MBPs), especially the 21.5-kDa isoform, but not the levels of total MBPs, decreased markedly during demyelination induced by aging, cuprizone treatment, and double knockout of FcRgamma/Fyn genes. We also showed that the recovery from demyelination in cuprizone-treated and aged mice is achieved after administration of the herbal medicine Ninjin'yoeito, an effective therapy targeting the FcRgamma/Fyn-Rho (Rac1)-MAPK (P38 MAPK)-p-MBPs signaling cascade. These results suggest that the restoration of FcRgamma/Fyn signaling represents a new approach for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Chika Seiwa
- Department of Neuro-Glia Cell Biology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zipp F, Aktas O. The brain as a target of inflammation: common pathways link inflammatory and neurodegenerative diseases. Trends Neurosci 2006; 29:518-27. [PMID: 16879881 DOI: 10.1016/j.tins.2006.07.006] [Citation(s) in RCA: 256] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Revised: 05/23/2006] [Accepted: 07/20/2006] [Indexed: 11/16/2022]
Abstract
Classical knowledge distinguishes between inflammatory and non-inflammatory diseases of the brain. Either the immune system acts on the CNS and initiates a damage cascade, as in autoimmune (e.g. multiple sclerosis) and infectious conditions, or the primary insult is not inflammation but ischemia or degeneration, as in stroke and Alzheimer's disease, respectively. However, as we review here, recent advances have blurred this distinction. On the one hand, the classical inflammatory diseases of the brain also exhibit profound and early neurodegenerative features - remarkably, it has been known for more than a century that neuronal damage is a key feature of multiple sclerosis pathology, yet this was neglected until very recently. On the other hand, immune mechanisms might set the pace of progressive CNS damage in primary neurodegeneration. Despite differing initial events, increasing evidence indicates that even in clinically heterogeneous diseases, there might be common immunological pathways that result in neurotoxicity and reveal targets for more efficient therapies.
Collapse
Affiliation(s)
- Frauke Zipp
- Institute of Neuroimmunology, Charité - Universitätsmedizin Berlin, 10098 Berlin, Germany.
| | | |
Collapse
|
38
|
Zhu KY, Feferman T, Maiti PK, Souroujon MC, Fuchs S. Intravenous immunoglobulin suppresses experimental myasthenia gravis: Immunological mechanisms. J Neuroimmunol 2006; 176:187-97. [PMID: 16730380 DOI: 10.1016/j.jneuroim.2006.04.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 03/21/2006] [Accepted: 04/10/2006] [Indexed: 11/16/2022]
Abstract
Intravenous immunoglobulin (IVIG) administration has been beneficially used in the treatment of several autoimmune disorders including myasthenia gravis (MG), although its mechanism of action is still not clear. To study the optimal conditions of IVIG treatment and delineate its mechanism of action we established a suitable model in rat experimental autoimmune MG (EAMG). We show that IVIG has a suppressive effect on the clinical symptoms of ongoing EAMG that is associated with decreased AChR-specific cellular and humoral immune reactivity. Costimulatory factors and cytokine profile analyses suggest that IVIG immunomodulation in EAMG involves suppression of B and Th1-type T cell responses with no generation of T-regulatory cells. Our data contribute to the understanding of the immunological mechanisms underlying IVIG treatment in MG and in other autoimmune disorders.
Collapse
Affiliation(s)
- Kai-Yun Zhu
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
39
|
Abstract
Intravenous immunoglobulin (IVIg) has been used as an immunomodulatory therapy for the treatment of multiple sclerosis (MS). In the current review, we summarize the up-to-date data related to IVIg clinical trials in MS, and the suggested mechanisms of action by which IVIg modulates the relevant immunological pathways impaired in MS.
Collapse
Affiliation(s)
- Anat Achiron
- Multiple Sclerosis Center, Sheba Medical Center, Tel-Hashomer, Israel.
| | | |
Collapse
|
40
|
Guilpain P, Chanseaud Y, Tamby MC, Larroche C, Guillevin L, Kaveri SV, Kazatchkine MD, Mouthon L. Effets immunomodulateurs des immunoglobulines intraveineuses. Presse Med 2004; 33:1183-94. [PMID: 15523290 DOI: 10.1016/s0755-4982(04)98888-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Intravenous immunoglobulins (IVIg) are therapeutic preparations of normal human IgG obtained from pools of more than 1000 healthy blood donors. They are currently used in the treatment of a wide range of auto-immune diseases, whether associated with auto-antibodies or auto-reactive T lymphocytes, as well as in the treatment of systemic inflammatory diseases. Several mechanisms of action have been identified during the last 20 years, including: (i) modulation of Fc receptors expression on leukocytes and endothelial cells; (ii) interaction with complement proteins; (iii) modulation of cytokines and chemokines synthesis and release; (iv) modulation of cell proliferation and apoptosis; (v) remyelinisation; (vi) neutralisation of circulating autoantibodies; (vii) selection of repertoires of B and T lymphocytes; (viii) interaction with other cell-surface molecules on lymphocytes and monocytes; (ix) corticosteroid sparing. These mechanisms of action are multiple and often intricate. However, they are still little known and further investigations are warranted.
Collapse
Affiliation(s)
- Philippe Guilpain
- Service de médecine interne, Hôpital Cochin, Assistance publique-Hôpitaux de Paris et Université Paris V, Paris
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Karnezis T, Mandemakers W, McQualter JL, Zheng B, Ho PP, Jordan KA, Murray BM, Barres B, Tessier-Lavigne M, Bernard CCA. The neurite outgrowth inhibitor Nogo A is involved in autoimmune-mediated demyelination. Nat Neurosci 2004; 7:736-44. [PMID: 15184901 DOI: 10.1038/nn1261] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2004] [Accepted: 04/29/2004] [Indexed: 11/08/2022]
Abstract
Inhibitors associated with CNS myelin are thought to be important in the failure of axons to regenerate after spinal cord injury and in other neurodegenerative disorders. Here we show that targeting the CNS-specific inhibitor of neurite outgrowth Nogo A by active immunization blunts clinical signs, demyelination and axonal damage associated with experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). Mice vaccinated against Nogo A produce Nogo-specific antibodies that block the neurite outgrowth inhibitory activity associated with CNS myelin in vitro. Passive immunization with anti-Nogo IgGs also suppresses EAE. Our results identify Nogo A as an important determinant of the development of EAE and suggest that its blockade may help to maintain and/or to restore the neuronal integrity of the CNS after autoimmune insult in diseases such as MS. Our finding that Nogo A is involved in CNS autoimmune demyelination indicates that this molecule may have a far more complex role than has been previously anticipated.
Collapse
Affiliation(s)
- Tara Karnezis
- Neuroimmunology Laboratory, Department of Biochemistry, School of Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Binstadt BA, Geha RS, Bonilla FA. IgG Fc receptor polymorphisms in human disease: implications for intravenous immunoglobulin therapy. J Allergy Clin Immunol 2003; 111:697-703. [PMID: 12704346 DOI: 10.1067/mai.2003.1380] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Polymorphisms of human Fc receptors (FcRs) have been described that are associated with the development or progression of autoimmune diseases. The FcR polymorphisms affect the affinity with which FcRs interact with immunoglobulin molecules. Intravenous immunoglobulin is administered as therapy for many autoimmune diseases and might exert its effects by interacting with FcRs. Thus, FcR polymorphisms might influence the efficacy of intravenous immunoglobulin therapy for patients with certain autoimmune diseases. In this article we review FcR polymorphisms in relation to autoimmune diseases for which intravenous immunoglobulin is used therapeutically.
Collapse
MESH Headings
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmune Diseases/therapy
- Granulomatosis with Polyangiitis/genetics
- Granulomatosis with Polyangiitis/immunology
- Granulomatosis with Polyangiitis/therapy
- Guillain-Barre Syndrome/genetics
- Guillain-Barre Syndrome/immunology
- Guillain-Barre Syndrome/therapy
- Humans
- Immunoglobulins, Intravenous/pharmacology
- Immunoglobulins, Intravenous/therapeutic use
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/therapy
- Polymorphism, Genetic
- Purpura, Thrombocytopenic, Idiopathic/genetics
- Purpura, Thrombocytopenic, Idiopathic/immunology
- Purpura, Thrombocytopenic, Idiopathic/therapy
- Receptors, IgG/genetics
Collapse
Affiliation(s)
- Bryce A Binstadt
- Division of Immunology, Children's Hospital Boston, MA 02115, USA
| | | | | |
Collapse
|
43
|
Aukrust P, Damås JK, Gullestad L. Immunomodulating therapy: new treatment modality in congestive heart failure. CONGESTIVE HEART FAILURE (GREENWICH, CONN.) 2003; 9:64-9. [PMID: 12671336 DOI: 10.1111/j.1527-5299.2003.00682.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Accumulating evidence indicates that inflammatory cytokines play a pathogenic role in congestive heart failure (CHF) by influencing heart contractility, inducing hypertrophy, and promoting apoptosis or fibrosis, contributing to the continuous myocardial remodeling process. Traditional cardiovascular drugs seem to have little influence on the overall cytokine network, and immunomodulatory therapy has emerged as a possible new treatment modality in CHF. Several animal studies have suggested that modulation of inflammatory cytokines may improve cardiac performance. The authors have recently demonstrated that intravenous immunoglobulin enhances the left ventricular ejection fraction in CHF patients, and that this is significantly correlated with anti-inflammatory effects of such therapy. While intravenous immunoglobulin is not necessarily the drug of choice, this study suggests a potential role for immunomodulatory therapy in CHF in addition to optimal cardiovascular treatment regimens. Further research will more precisely identify the most important actors in the immunopathogenesis of CHF and contribute to the development of more specific immunomodulating agents for this disorder.
Collapse
Affiliation(s)
- Pål Aukrust
- Section of Clinical Immunology and Infectious Diseases, Medical Department, Rikshospitalet, Sognsvannsveien 20, 0027 Oslo, Norway.
| | | | | |
Collapse
|
44
|
Pul R, Nguyen D, Schmitz U, Marx P, Stangel M. Comparison of intravenous immunoglobulin preparations on microglial function in vitro: more potent immunomodulatory capacity of an IgM/IgA-enriched preparation. Clin Neuropharmacol 2002; 25:254-9. [PMID: 12410056 DOI: 10.1097/00002826-200209000-00005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Intravenous immunoglobulins (IVIg) have been used successfully as an immunomodulating treatment for patients with inflammatory diseases of the central nervous system (CNS) including multiple sclerosis (MS). It was shown previously that IVIg could modulate the functions of microglia, the main immune cell in the CNS. We have compared five commercially available IVIg preparations on their capacity to modulate tumor necrosis factor (TNF)-alpha secretion and nitric oxide production in cultured microglia. All preparations induced a dose-dependent stimulation of TNF-alpha secretion as measured by ELISA. There were some small differences between preparations consisting of IgG, while the preparation enriched for IgM and IgA induced a considerably higher TNF-alpha production at 1 mg/mL and 10 mg/mL. Similar results were seen for nitric oxide production as measured indirectly by the Griess reaction. These results indicate that IgM/IgA-enriched IVIg may be a more potent immunomodulator than pure IgG preparations on inflammatory reactions in the CNS.
Collapse
Affiliation(s)
- Refik Pul
- Department of Neurology, Universtätsklinikum Benjamin Franklin, Freie Universität Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
45
|
Larroche C, Chanseaud Y, Garcia de la Pena-Lefebvre P, Mouthon L. Mechanisms of intravenous immunoglobulin action in the treatment of autoimmune disorders. BioDrugs 2002; 16:47-55. [PMID: 11909001 DOI: 10.2165/00063030-200216010-00005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Intravenous immunoglobulins (IVIg) are therapeutic preparations of normal human immunoglobulin (Ig) G obtained from pools of blood from more than 1000 healthy donors, and exert immunomodulatory effects in autoantibody-mediated and T-cell-mediated autoimmune disorders and systemic inflammatory diseases. IVIg mechanisms of action in autoimmune diseases have been extensively analysed during the last 15 years and include the following: (i) interaction of the IgG Fc fragment with Fc receptors on leucocytes and endothelial cells; (ii) interaction of infused IgG with complement proteins; (iii) monocyte and lymphocyte modulation of synthesis and release of cytokines and cytokine antagonists; (iv) modulation of cell proliferation and reparation; (v) neutralisation of circulating autoantibodies; (vi) selection of immune repertoires; and (vii) interaction with other cell-surface molecules on T and B lymphocytes.
Collapse
Affiliation(s)
- Claire Larroche
- Department of Internal Medicine and Infectious Diseases, Hôpital Avicenne and Assistance Publique-Hôpitaux de Paris, 125 rue de Stalingrad, Bobigny Cedex 93009, France
| | | | | | | |
Collapse
|
46
|
Lock C, Hermans G, Pedotti R, Brendolan A, Schadt E, Garren H, Langer-Gould A, Strober S, Cannella B, Allard J, Klonowski P, Austin A, Lad N, Kaminski N, Galli SJ, Oksenberg JR, Raine CS, Heller R, Steinman L. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med 2002; 8:500-8. [PMID: 11984595 DOI: 10.1038/nm0502-500] [Citation(s) in RCA: 1277] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microarray analysis of multiple sclerosis (MS) lesions obtained at autopsy revealed increased transcripts of genes encoding inflammatory cytokines, particularly interleukin-6 and -17, interferon-gamma and associated downstream pathways. Comparison of two poles of MS pathology--acute lesions with inflammation versus 'silent' lesions without inflammation--revealed differentially transcribed genes. Some products of these genes were chosen as targets for therapy of experimental autoimmune encephalomyelitis (EAE) in mice. Granulocyte colony-stimulating factor is upregulated in acute, but not in chronic, MS lesions, and the effect on ameliorating EAE is more pronounced in the acute phase, in contrast to knocking out the immunoglobulin Fc receptor common gamma chain where the effect is greatest on chronic disease. These results in EAE corroborate the microarray studies on MS lesions. Large-scale analysis of transcripts in MS lesions elucidates new aspects of pathology and opens possibilities for therapy.
Collapse
Affiliation(s)
- Christopher Lock
- Department of Neurology and Neurological Sciences, Beckman Center, Stanford University, Stanford, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Keren G, Keren P, Barshack I, Pri-Chen S, George J. The effect of intravenous immunoglobulins on intimal thickening in a mouse model of arterial injury. Atherosclerosis 2001; 159:77-83. [PMID: 11689209 DOI: 10.1016/s0021-9150(01)00491-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Inflammatory mechanisms appear to influence the progression of intimal thickening in experimental models of arterial injury. Intravenous immunoglobulin (IVIG) is a polyspecific preparation of human immunoglobulin (Ig)G employed for treatment of autoimmune disorders. In this study, we sought to investigate whether treatment with IVIG could influence intimal thickening in a model of murine arterial injury. Intimal thickening was induced by placement of a periadventitial cuff over the right femoral artery of male C57BL/6 mice. In the first experiment, IVIG or human serum albumin (HSA) (10 mg/mouse) were administered intraperitoneally for five consecutive days starting 1 day prior to cuff placement. In the second experiment, IVIG or HSA treatment were delivered similarly, but initiated 3 days following induction of arterial injury. Neointimal area and intimal/medial ratio were significantly reduced in mice treated with IVIG prior to cuff placement as compared with HSA treatment. No differences were noted with regard to neointimal area or intimal/medial ratio, between IVIG- and HSA-treated mice when the treatment was commenced 3 days following induction of injury. IVIG treatment reduced the proliferative capacity of splenocytes to the non-specific mitogen Con-A. Treatment with IVIG was associated with a significantly enhanced secretion of interleukin (IL)-10) by the respective splenocytes in comparison with HSA-treated mice. No effect of IVIG was evident on the secretion of IL-4 or IFN-gamma. Thus, IVIG has proven beneficial in ameliorating intimal thickening in a mouse model of arterial injury. The effect could be mediated by upregulation of T-cell secretion of the anti-inflammatory cytokine IL-10.
Collapse
Affiliation(s)
- G Keren
- Department of Cardiology and the Cardiovascular Research Laboratory, Ichilov Hospital, Elias Sourasky Tel-Aviv Medical Center, 6 Weizman Street, Tel-Aviv, Israel.
| | | | | | | | | |
Collapse
|
48
|
George J, Barshack I, Malka E, Goldberg I, Keren P, Laniado S, Keren G. The effect of intravenous immunoglobulins on the progression of experimental autoimmune myocarditis in the rat. Exp Mol Pathol 2001; 71:55-62. [PMID: 11502097 DOI: 10.1006/exmp.2001.2382] [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: 11/22/2022]
Abstract
The aim of this study was to evaluate the efficacy of two experimental regimes of human intravenous immunoglobulins (IVIG) on the progression of experimental autoimmune myocarditis (EAM). EAM is induced by immunization against myosin and represents a T-cell-dependent disorder that progresses toward dilated cardiomyopathy similar to the human equivalent. No effective treatment is currently at hand for management of the disorder, as immunosuppressant drugs are associated with multiple side effects. Three groups of Lewis rats were induced to develop EAM by immunization with porcine myosin and sacrificed 21 days later. Group A received a 5-day regimen of IVIG (800 mg/kg) following induction of the disorder; Group B received a daily dose of IVIG (800 mg/kg) and group C was treated with PBS. IVIG given daily but not during the first 5 days significantly suppressed myocarditis score (0.81 +/- 0.26 and 1.14 +/- 0.42, respectively) in comparison with controls (mean score of 1.78 +/- 0.36). The effect was accompanied by a reduction in the cellular and humoral immune response of the respective animals toward myosin. IVIG was deposited within the extracellular matrix surrounding the damaged myocytes. TNF-alpha expression was reduced in both groups treated with IVIG, whereas iNOS expression paralleled the extent of myocardial inflammation regardless of treatment. IVIG at doses twice those applied for human disease are effective in ameliorating the progression of EAM. The effect may be mediated by suppression of the cellular and humoral response to myosin. IVIG may be found clinically feasible in humans as an adjuvant or single therapy for autoimmune myocarditis.
Collapse
Affiliation(s)
- J George
- Department of Cardiology and Cardiovascular Research Laboratory, Tel-Aviv Medical Center, Tel-Aviv, Israel
| | | | | | | | | | | | | |
Collapse
|
49
|
Aktas O, Waiczies S, Grieger U, Wendling U, Zschenderlein R, Zipp F. Polyspecific immunoglobulins (IVIg) suppress proliferation of human (auto)antigen-specific T cells without inducing apoptosis. J Neuroimmunol 2001; 114:160-7. [PMID: 11240027 DOI: 10.1016/s0165-5728(01)00243-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Polyspecific immunoglobulins (IVIg) have been shown to reduce disease activity in multiple sclerosis (MS). To investigate the mechanisms of action of IVIg, we studied the impact of IVIg on growth and death (apoptosis) of human (auto)antigen-specific T cells. We observed a substantial suppression of proliferation of specifically activated T cells, in absence of caspase activation or DNA fragmentation. Further, neither susceptibility of T cells to undergo CD95-mediated apoptosis nor expression of apoptosis-blocking bcl-2 was modulated by IVIg. We conclude that IVIg may inhibit the reactivity of antigen-specific T cells in MS through suppression of proliferation rather than modulation of apoptosis.
Collapse
Affiliation(s)
- O Aktas
- Department of Neurology, Division of Neuroimmunology, University Hospital Charité, NWFZ 2680, Schumannstrasse 20/21, 10117 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
50
|
LeClaire RD, Bavari S. Human antibodies to bacterial superantigens and their ability to inhibit T-cell activation and lethality. Antimicrob Agents Chemother 2001; 45:460-3. [PMID: 11158741 PMCID: PMC90313 DOI: 10.1128/aac.45.2.460-463.2001] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial superantigens (BSAgs) cause massive stimulation of the immune system and are associated with various pathologies and diseases. To address the role of antibodies in protection against BSAgs, we screened the sera of 29 human volunteers for antibodies to the SAgs staphylococcal enterotoxin A (SEA), SEB, SEC1, and toxic shock syndrome toxin 1 (TSST-1). Although all volunteers had detectable levels of antibodies against SEB and SEC1, many (9 out of 29 volunteers) lacked detectable antibody to SEA or had minimal titers. Antibody titers to TSST-1 were well below those to SEB and SEC1, and three volunteers lacked detectable antibody to this BSAg. In addition, pooled immunoglobulin preparations obtained from different companies had antibody titers against SEs and TSST-1. There was a good correlation between antibody titers and inhibition of superantigenic effects of these toxins. Transfer of SEB-specific antibodies, obtained from pooled sera, suppressed in vitro T-cell proliferation and totally protected mice against SEB. These data suggest that the inhibitory activity of human sera was specific to antibodies directed against the toxins. Thus, it may be possible to counteract with specific antibodies BSAg-associated pathologies caused by stimulation of the immune system.
Collapse
Affiliation(s)
- R D LeClaire
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702-5011, USA
| | | |
Collapse
|