1
|
Wu J, Yang H, Yu D, Yang X. Blood-derived product therapies for SARS-CoV-2 infection and long COVID. MedComm (Beijing) 2023; 4:e426. [PMID: 38020714 PMCID: PMC10651828 DOI: 10.1002/mco2.426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is capable of large-scale transmission and has caused the coronavirus disease 2019 (COVID-19) pandemic. Patients with COVID-19 may experience persistent long-term health issues, known as long COVID. Both acute SARS-CoV-2 infection and long COVID have resulted in persistent negative impacts on global public health. The effective application and development of blood-derived products are important strategies to combat the serious damage caused by COVID-19. Since the emergence of COVID-19, various blood-derived products that target or do not target SARS-CoV-2 have been investigated for therapeutic applications. SARS-CoV-2-targeting blood-derived products, including COVID-19 convalescent plasma, COVID-19 hyperimmune globulin, and recombinant anti-SARS-CoV-2 neutralizing immunoglobulin G, are virus-targeting and can provide immediate control of viral infection in the short term. Non-SARS-CoV-2-targeting blood-derived products, including intravenous immunoglobulin and human serum albumin exhibit anti-inflammatory, immunomodulatory, antioxidant, and anticoagulatory properties. Rational use of these products can be beneficial to patients with SARS-CoV-2 infection or long COVID. With evidence accumulated since the pandemic began, we here summarize the progress of blood-derived product therapies for COVID-19, discuss the effective methods and scenarios regarding these therapies, and provide guidance and suggestions for clinical treatment.
Collapse
Affiliation(s)
- Junzheng Wu
- Chengdu Rongsheng Pharmaceuticals Co., Ltd.ChengduChina
| | | | - Ding Yu
- Chengdu Rongsheng Pharmaceuticals Co., Ltd.ChengduChina
- Beijing Tiantan Biological Products Co., Ltd.BeijingChina
| | | |
Collapse
|
2
|
Liu Y, Tong H, He F, Zhai Y, Wu C, Wang J, Jiang C. Effect of intravenous immunoglobulin therapy on the prognosis of patients with severe fever with thrombocytopenia syndrome and neurological complications. Front Immunol 2023; 14:1118039. [PMID: 37033957 PMCID: PMC10073413 DOI: 10.3389/fimmu.2023.1118039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/08/2023] [Indexed: 04/11/2023] Open
Abstract
Background Intravenous immunoglobulin (IVIG) has been reported to exert a beneficial effect on severe fever with thrombocytopenia syndrome (SFTS) patients with neurological complications. However, in clinical practice, the standard regime is unclear and there is a lack of evidence from large-scale studies. Methods A single-center retrospective study was conducted to determine the influence of IVIG dosage and duration on SFTS patients with neurological complications. The primary outcome was 28-day mortality, and laboratory parameters before and after IVIG treatment were measured. Survival curves were generated using the Kaplan-Meier method and analyzed with the log-rank test according to the median IVIG dosage and IVIG duration. Besides, multivariate Cox regression analysis was performed to examine the association between the independent factors and 28-day mortality in SFTS patients. Results Overall, 36 patients (58.06%) survived, while 26 (41.9%) patients died. The median age of the included patients was 70 (55-75) years, and 46.8% (29/62) were male. A significantly higher clinical presentation of dizziness and headache was observed in the survival group. The IVIG duration in the survival group was longer than in the death group (P <0.05). Additionally, the IVIG dosage was higher in the survival group than in the death group, but there was not a statistically significant difference between the two groups (P = 0.066). The mediating effect of IVIG duration was verified through the relationship between IVIG dosage and prognosis using the Sobel test. Univariate analysis revealed that IVIG dosage (HR: 0.98; 95% CI: 0.97-1.00; P = 0.007) and IVIG duration (HR: 0.54; 95% CI: 0.41-0.72; P <0.001) were significantly associated with risk of death. The multivariate analysis generated an adjusted HR value of 0.98 (95% CI: 0.96-1.00; P = 0.012) for IVIG dosage and 0.26 (95% CI: 0.09-0.78; P = 0.016) for dizziness and headache. Conclusion Prolonged high-dose IVIG is beneficial to the 28-day prognosis in SFTS patients with neurological complications.
Collapse
Affiliation(s)
- Yun Liu
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hanwen Tong
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fei He
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yu Zhai
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chao Wu
- Department of Infectious Disease, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jun Wang
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chenxiao Jiang
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
3
|
Fei X, Fang K, Ni X, Ren WH. Risk Factors of Neurological Complications in Severe Fever Patients with Thrombolytic Syndrome: A Single-Center Retrospective Study in China. Med Sci Monit 2021; 27:e932836. [PMID: 34744159 PMCID: PMC8588710 DOI: 10.12659/msm.932836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Severe fever with thrombocytopenia syndrome is a serious insect-borne infectious disease caused by the Huaiyangshanbanyang virus. We conducted a retrospective study to identify risk factors for neurological complications caused by the virus. Material/Methods We included 121 patients who had severe fever with thrombocytopenia syndrome and were admitted to our hospital from 2013 to 2020. Patients’ laboratory test results and clinical data were collected. Univariate and multivariate regression were used for statistical analysis. Results Patients with neurological complications had higher mortality rates and longer hospital stays and disease duration than did patients without neurological complications. The neurological symptoms with the highest incidence rates were involuntary tremors (tongue and mandible), cognitive disorder, and limb tremors. Patients with neurological complications had a higher incidence of abnormal heart rhythms. Subcutaneous bleeding, pulmonary rales, percentage of neutrophils, increased lactate dehydrogenase and C-reactive protein levels, and decreased chloride ion concentration were closely related to the occurrence of neurological complications. The significant decrease in chloride ion concentration within 1 to 5 days of disease onset may be a risk factor for predicting the occurrence of neurological complications in patients with severe fever with thrombocytopenia syndrome. Conclusions Early monitoring of subcutaneous bleeding, pulmonary rales, electrocardiogram changes, and biochemical indicators in patients with severe fever with thrombocytopenia syndrome can predict the occurrence of neurological complications.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Infectious Diseases, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland).,Department of Infectious Diseases, Yidu Central Hospital of Weifang Affiliated to Weifang Medical University, Weifang, Shandong, China (mainland)
| | - Kai Fang
- Department of Vertigo Medicine, Qingzhou Hospital Affiliated to Shandong First Medical University, Weifang, Shandong, China (mainland)
| | - Xiuying Ni
- Department of Infectious Diseases, Yidu Central Hospital of Weifang Affiliated to Weifang Medical University, Weifang, Shandong, China (mainland)
| | - Wan-Hua Ren
- Department of Infectious Diseases, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
4
|
Li X, Tian DC, Fan M, Xiu Y, Wang X, Li T, Jia D, Xu W, Song T, Shi FD, Zhang X. Intravenous immunoglobulin for acute attacks in neuromyelitis optica spectrum disorders (NMOSD). Mult Scler Relat Disord 2020; 44:102325. [DOI: 10.1016/j.msard.2020.102325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/07/2020] [Accepted: 06/21/2020] [Indexed: 10/24/2022]
|
5
|
Groh J, Martini R. Neuroinflammation as modifier of genetically caused neurological disorders of the central nervous system: Understanding pathogenesis and chances for treatment. Glia 2017; 65:1407-1422. [PMID: 28568966 DOI: 10.1002/glia.23162] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 12/21/2022]
Abstract
Genetically caused neurological disorders of the central nervous system (CNS) are usually orphan diseases with poor or even fatal clinical outcome and few or no treatments that will improve longevity or at least quality of life. Neuroinflammation is common to many of these disorders, despite the fact that a plethora of distinct mutations and molecular changes underlie the disorders. In this article, data from corresponding animal models are analyzed to define the roles of innate and adaptive inflammation as modifiers and amplifiers of disease. We describe both common and distinct patterns of neuroinflammation in genetically mediated CNS disorders and discuss the contrasting mechanisms that lead to adverse versus neuroprotective effects. Moreover, we identify the juxtaparanode as a neuroanatomical compartment commonly associated with inflammatory cells and ongoing axonopathic changes, in models of diverse diseases. The identification of key immunological effector pathways that amplify neuropathic features should lead to realistic possibilities for translatable therapeutic interventions using existing immunomodulators. Moreover, evidence emerges that neuroinflammation is not only able to modify primary neural damage-related symptoms but also may lead to unexpected clinical outcomes such as neuropsychiatric syndromes.
Collapse
Affiliation(s)
- Janos Groh
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str. 11, Würzburg, D-97080, Germany
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str. 11, Würzburg, D-97080, Germany
| |
Collapse
|
6
|
Maddur MS, Stephen-Victor E, Das M, Prakhar P, Sharma VK, Singh V, Rabin M, Trinath J, Balaji KN, Bolgert F, Vallat JM, Magy L, Kaveri SV, Bayry J. Regulatory T cell frequency, but not plasma IL-33 levels, represents potential immunological biomarker to predict clinical response to intravenous immunoglobulin therapy. J Neuroinflammation 2017; 14:58. [PMID: 28320438 PMCID: PMC5360043 DOI: 10.1186/s12974-017-0818-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/18/2017] [Indexed: 02/08/2023] Open
Abstract
Background Intravenous immunoglobulin (IVIG) is a polyspecific pooled immunoglobulin G preparation and one of the commonly used therapeutics for autoimmune diseases including those of neurological origin. A recent report in murine model proposed that IVIG expands regulatory T (Treg) cells via induction of interleukin 33 (IL-33). However, translational insight on these observations is lacking. Methods Ten newly diagnosed Guillain-Barré syndrome (GBS) patients were treated with IVIG at the rate of 0.4 g/kg for three to five consecutive days. Clinical evaluation for muscular weakness was performed by Medical Research Council (MRC) and modified Rankin scoring (MRS) system. Heparinized blood samples were collected before and 1, 2, and 4–5 weeks post-IVIG therapy. Peripheral blood mononuclear cells were stained for surface CD4 and intracellular Foxp3, IFN-γ, and tumor necrosis factor alpha (TNF-α) and were analyzed by flow cytometry. IL-33 and prostaglandin E2 in the plasma were measured by ELISA. Results The fold changes in plasma IL-33 at week 1 showed no correlation with the MRC and MRS scores at weeks 1, 2, and ≥4 post-IVIG therapy. Clinical recovery following IVIG therapy appears to be associated with Treg cell response. Contrary to murine study, there was no association between the fold changes in IL-33 at week 1 and Treg cell frequency at weeks 1, 2, and ≥4 post-IVIG therapy. Treg cell-mediated clinical response to IVIG therapy in GBS patients was associated with reciprocal regulation of effector T cells-expressing TNF-α. Conclusion Treg cell expansion by IVIG in patients with autoimmune diseases lack correlation with IL-33. Treg cell frequency, but not plasma IL-33 levels, represents potential immunological biomarker to predict clinical response to IVIG therapy.
Collapse
Affiliation(s)
- Mohan S Maddur
- Institut National de la Santé et de la Recherche Médicale, Unité 1138, Paris, 75006, France.,Centre de Recherche des Cordeliers, Equipe- Immunopathologie et immuno-intervention thérapeutique, Paris, 75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, 75006, France.,Université Paris Descartes, UMR S 1138, Paris, 75006, France.,Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Emmanuel Stephen-Victor
- Institut National de la Santé et de la Recherche Médicale, Unité 1138, Paris, 75006, France.,Centre de Recherche des Cordeliers, Equipe- Immunopathologie et immuno-intervention thérapeutique, Paris, 75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, 75006, France
| | - Mrinmoy Das
- Institut National de la Santé et de la Recherche Médicale, Unité 1138, Paris, 75006, France.,Centre de Recherche des Cordeliers, Equipe- Immunopathologie et immuno-intervention thérapeutique, Paris, 75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, 75006, France
| | - Praveen Prakhar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
| | - Varun K Sharma
- Institut National de la Santé et de la Recherche Médicale, Unité 1138, Paris, 75006, France.,Centre de Recherche des Cordeliers, Equipe- Immunopathologie et immuno-intervention thérapeutique, Paris, 75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, 75006, France.,Université Paris Descartes, UMR S 1138, Paris, 75006, France
| | - Vikas Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
| | - Magalie Rabin
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, 75006, France
| | - Jamma Trinath
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
| | - Kithiganahalli N Balaji
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
| | - Francis Bolgert
- Réanimation Neurologique, Neurologie 1, Hôpital de la Pitié-Salpêtrière, Paris, 75651, France
| | - Jean-Michel Vallat
- Centre de Référence 'Neuropathies Périphériques Rares' et Service de Neurologie, Hôpital Universitaire Limoges, Limoges, 87042, France
| | - Laurent Magy
- Centre de Référence 'Neuropathies Périphériques Rares' et Service de Neurologie, Hôpital Universitaire Limoges, Limoges, 87042, France
| | - Srini V Kaveri
- Institut National de la Santé et de la Recherche Médicale, Unité 1138, Paris, 75006, France. .,Centre de Recherche des Cordeliers, Equipe- Immunopathologie et immuno-intervention thérapeutique, Paris, 75006, France. .,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, 75006, France. .,Université Paris Descartes, UMR S 1138, Paris, 75006, France.
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Unité 1138, Paris, 75006, France. .,Centre de Recherche des Cordeliers, Equipe- Immunopathologie et immuno-intervention thérapeutique, Paris, 75006, France. .,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, 75006, France. .,Université Paris Descartes, UMR S 1138, Paris, 75006, France.
| |
Collapse
|
7
|
Totzeck A, Stettner M, Hagenacker T. Early platelet and leukocyte decline in patients with neuroinflammatory disorders after intravenous immunoglobulins. Eur J Neurol 2017; 24:638-644. [PMID: 28224702 DOI: 10.1111/ene.13264] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/11/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE Intravenous immunoglobulins (IVIGs) are a common therapy in patients with neuroinflammatory disorders, especially chronic inflammatory demyelinating polyradiculoneuropathy or Guillain-Barré syndrome. Hematological toxicities upon IVIG infusion are a known side effect and still an important subject of investigation. METHODS Laboratory results and data for clinical efficacy and tolerability of 62 patients with neuroinflammatory disorders treated with IVIG (0.4 g/kg bodyweight per day over 5 days) at the Department of Neurology, University of Duisburg-Essen, Germany, were retrospectively analyzed. Blood samples were taken before and 1 day after IVIG administration. RESULTS In pre-treated and first-time treated patients, there was a significant decrease in white blood cell count (WBC) (8.10 ± 2.85/nl to 5.61 ± 2.50/nl, P < 0.001, n = 57) and platelets (255 ± 72/nl to 215 ± 66/nl, P < 0.001, n = 57). Mild hemolysis of red blood cells was found in patients who received IVIG for the first time (red blood cell count 4.61 ± 0.67/pl to 4.28 ± 0.52/pl, hemoglobin 13.7 ± 1.7 g/l to 13.0 ± 1.7 g/l, P < 0.001, n = 40). Hemolysis was associated with less tolerability of IVIG treatment and clinical efficacy was accompanied with a higher decline of WBC (not significant). CONCLUSIONS Next to mild hemolysis, a significant decrease in WBC and platelets can be detected early after high dose IVIGs in patients with neuroinflammatory disorders. Changes in blood counts may be possible markers for clinical efficacy and tolerability. Patients with low blood counts in advance should be particularly closely monitored whilst on IVIG treatment.
Collapse
Affiliation(s)
- A Totzeck
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - M Stettner
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - T Hagenacker
- Department of Neurology, University Hospital Essen, Essen, Germany
| |
Collapse
|
8
|
Winter M, Baksmeier C, Steckel J, Barman S, Malviya M, Harrer-Kuster M, Hartung HP, Goebels N. Dose-dependent inhibition of demyelination and microglia activation by IVIG. Ann Clin Transl Neurol 2016; 3:828-843. [PMID: 27844029 PMCID: PMC5099529 DOI: 10.1002/acn3.326] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 04/25/2016] [Accepted: 05/09/2016] [Indexed: 12/31/2022] Open
Abstract
Objective Intravenous immunoglobulin (IVIG) is an established treatment for numerous autoimmune conditions. Clinical trials of IVIG for multiple sclerosis, using diverse dose regimens, yielded controversial results. The aim of this study is to dissect IVIG effector mechanisms on demyelination in an ex vivo model of the central nervous system (CNS)‐immune interface. Methods Using organotypic cerebellar slice cultures (OSC) from transgenic mice expressing green fluorescent protein (GFP) in oligodendrocytes/myelin, we induced extensive immune‐mediated demyelination and oligodendrocyte loss with an antibody specific for myelin oligodendrocyte glycoprotein (MOG) and complement. Protective IVIG effects were assessed by live imaging of GFP expression, confocal microscopy, immunohistochemistry, gene expression analysis and flow cytometry. Results IVIG protected OSC from demyelination in a dose‐dependent manner, which was at least partly attributed to interference with complement‐mediated oligodendroglia damage, while binding of the anti‐MOG antibody was not prevented. Staining with anti‐CD68 antibodies and flow cytometry confirmed that IVIG prevented microglia activation and oligodendrocyte death, respectively. Equimolar IVIG‐derived Fab fragments or monoclonal IgG did not protect OSC, while Fc fragments derived from a polyclonal mixture of human IgG were at least as potent as intact IVIG. Interpretation Both intact IVIG and Fc fragments exert a dose‐dependent protective effect on antibody‐mediated CNS demyelination and microglia activation by interfering with the complement cascade and, presumably, interacting with local immune cells. Although this experimental model lacks blood–brain barrier and peripheral immune components, our findings warrant further studies on optimal dose finding and alternative modes of application to enhance local IVIG concentrations at the site of tissue damage.
Collapse
Affiliation(s)
- Meike Winter
- Department of Neurology, Medical Faculty Heinrich-Heine-University Duesseldorf Moorenstr. 5 D-40225 Duesseldorf Germany
| | - Christine Baksmeier
- Department of Neurology, Medical Faculty Heinrich-Heine-University Duesseldorf Moorenstr. 5 D-40225 Duesseldorf Germany
| | - Julia Steckel
- Department of Neurology, Medical Faculty Heinrich-Heine-University Duesseldorf Moorenstr. 5 D-40225 Duesseldorf Germany
| | - Sumanta Barman
- Department of Neurology, Medical Faculty Heinrich-Heine-University Duesseldorf Moorenstr. 5 D-40225 Duesseldorf Germany
| | - Manish Malviya
- Department of Neurology, Medical Faculty Heinrich-Heine-University Duesseldorf Moorenstr. 5D-40225 Duesseldorf Germany; Present address: CPTP, Centre Physiopathologie de Toulouse-Purpan INSERM U1043 - CNRS UMR 5282-Université Toulouse III Toulouse France
| | - Melanie Harrer-Kuster
- University of Zuerich, Clinical Neuroimmunology Zuerich Switzerland; Present address: Abb Vie AG Baar Switzerland
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty Heinrich-Heine-University Duesseldorf Moorenstr. 5 D-40225 Duesseldorf Germany
| | - Norbert Goebels
- Department of Neurology, Medical Faculty Heinrich-Heine-University Duesseldorf Moorenstr. 5 D-40225 Duesseldorf Germany
| |
Collapse
|
9
|
Frigo G, Tramentozzi E, Orso G, Ceolotto G, Pagetta A, Stagni C, Menin C, Rosato A, Finotti P. Human IgGs induce synthesis and secretion of IgGs and neonatal Fc receptor in human umbilical vein endothelial cells. Immunobiology 2016; 221:1329-1342. [PMID: 27523744 DOI: 10.1016/j.imbio.2016.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/16/2016] [Accepted: 08/04/2016] [Indexed: 11/17/2022]
Abstract
Human IgGs are increasingly used in the therapy of many different immune and inflammatory diseases, however their mechanism of action still remains unclear in most diseases. To gain insight into the mechanism by which IgGs might also exert their effects on endothelial cells, we tested human IgGs on human umbilical vein endothelial cells (HUVECs). IgGs induced a time-dependent increase in the synthesis and secretion of IgGs, together with a marked angiogenic-like transformation of HUVECs that was maximal after a 20-h incubation. IgGs stimulated IG gene transcription without affecting the process of gene rearrangement, already present in control HUVECs. The mechanism involved the activation of transcription factors with the increased expression of HSP90, HSP70 and inactive MMP-9 responsible for the phenotypic differentiation associated with the most intense IgG synthesis and secretion. However, even a short incubation with IgGs followed by recovery of cells was sufficient to trigger and sustain in time the synthesis and secretion of new IgGs, independently of the angiogenic-like transformation visible only when cells were continuously exposed to IgGs. Under the stimulus of IgGs, specific secretory pathways were also activated in HUVECs together with the expression of FcRn, which was always associated with IgGs of new synthesis, forming complexes that were also secreted. Our results disclose a so far unknown and unexpected mechanism of IgGs on HUVECs that behave as Ig-producing immune cells. Results might have relevance for the effects that IgGs also exert in vivo in physiological conditions.
Collapse
Affiliation(s)
- Giulia Frigo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy
| | - Elisa Tramentozzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy
| | - Genny Orso
- Scientific Institute, IRCCS Eugenio Medea, Conegliano, Treviso, Italy
| | - Giulio Ceolotto
- Department of Medicine, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Andrea Pagetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy
| | - Camilla Stagni
- Department of Surgery Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, Padua, Italy
| | - Chiara Menin
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Via Gattamelata 64, Padua, Italy
| | - Antonio Rosato
- Department of Surgery Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, Padua, Italy; Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, Via Gattamelata 64, Padua, Italy.
| | - Paola Finotti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Pharmacology Building, L.go E. Meneghetti 2, 35131 Padua, Italy.
| |
Collapse
|
10
|
Galeotti C, Hegde P, Das M, Stephen-Victor E, Canale F, Muñoz M, Sharma VK, Dimitrov JD, Kaveri SV, Bayry J. Heme oxygenase-1 is dispensable for the anti-inflammatory activity of intravenous immunoglobulin. Sci Rep 2016; 6:19592. [PMID: 26796539 PMCID: PMC4726216 DOI: 10.1038/srep19592] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023] Open
Abstract
Intravenous immunoglobulin G (IVIG) is used in the therapy of various autoimmune and inflammatory conditions. The mechanisms by which IVIG exerts anti-inflammatory effects are not completely understood. IVIG interacts with numerous components of the immune system including dendritic cells, macrophages, T and B cells and modulate their functions. Recent studies have reported that heme oxygenase-1 (HO-1) pathway plays an important role in the regulation of inflammatory response in several pathologies. Several therapeutic agents exert anti-inflammatory effects via induction of HO-1. Therefore, we aimed at exploring if anti-inflammatory effects of IVIG are mediated via HO-1 pathway. Confirming the previous reports, we report that IVIG exerts anti-inflammatory effects on innate cells as shown by the inhibitory effects on IL-6 and nitric oxide production and confers protection in experimental autoimmune encephalomyelitis (EAE) model. However, these effects were not associated with an induction of HO-1 either in innate cells such as monocytes, dendritic cells and macrophages or in the kidneys and liver of IVIG-treated EAE mice. Also, inhibition of endogenous HO-1 did not modify anti-inflammatory effects of IVIG. These results thus indicate that IVIG exerts anti-inflammatory effects independent of HO-1 pathway.
Collapse
Affiliation(s)
- Caroline Galeotti
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France.,Department of Pediatric Rheumatology, National Referral Centre of Auto-inflammatory Diseases, CHU de Bicêtre, le Kremlin Bicêtre, F-94270, France
| | - Pushpa Hegde
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France
| | - Mrinmoy Das
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France
| | - Emmanuel Stephen-Victor
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France
| | - Fernando Canale
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France
| | - Marcos Muñoz
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France
| | - Varun K Sharma
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France
| | - Jordan D Dimitrov
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Paris, F-75006, France
| | - Srini V Kaveri
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Paris, F-75006, France.,International Associated Laboratory IMPACT (Institut National de la Santé et de la Recherche Médicale, France - Indian council of Medical Research, India), National Institute of Immunohaematology, Mumbai, 400012, India
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, F-75006, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR S 1138, Paris, F-75006, France.,Centre de Recherche des Cordeliers, Equipe - Immunopathology and therapeutic immunointervention, Paris, F-75006, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Paris, F-75006, France.,International Associated Laboratory IMPACT (Institut National de la Santé et de la Recherche Médicale, France - Indian council of Medical Research, India), National Institute of Immunohaematology, Mumbai, 400012, India
| |
Collapse
|
11
|
Ritter C, Bobylev I, Lehmann HC. Chronic inflammatory demyelinating polyneuropathy (CIDP): change of serum IgG dimer levels during treatment with intravenous immunoglobulins. J Neuroinflammation 2015; 12:148. [PMID: 26268846 PMCID: PMC4535537 DOI: 10.1186/s12974-015-0361-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/09/2015] [Indexed: 12/31/2022] Open
Abstract
Background Intravenous immunoglobulin (IVIg) is an effective treatment in chronic inflammatory demyelinating polyneuropathy (CIDP). In most patients, the optimal IVIg dose and regime is unknown. Polyvalent immunoglobulin (Ig) G form idiotypic/anti-idiotypic antibody pairs in serum and IVIg preparations. We determined IgG dimer levels before and after IVIg treatment in CIDP patients with the aim to explore their utility to serve as a surrogate marker for treatment response. Methods IgG was purified from serum of five controls without treatment, as well as from serum of 16 CIDP patients, two patients with Miller Fisher syndrome (MFS), and one patient with myasthenia gravis before and after treatment with IVIg. IgG dimer levels were determined by size exclusion chromatography. IgG dimer formation was correlated with clinical response to IVIg treatment in CIDP. Re-monomerized IgG dimer fractions were analyzed for immunoreactivity against peripheral nerve tissue. Results IgG dimer levels were significantly higher in post- compared to pre-IVIg infusion samples. Low post-treatment IgG dimer levels in CIDP patients were associated with clinical worsening during IVIg treatment. Re-monomerized IgG dimer fractions from CIDP patients showed immunoreactivity against peripheral nerve tissue, whereas similarly treated samples from MFS patients showed immunoreactivity against GQ1b. Conclusion Assessment of IgG dimer levels could be a novel approach to monitor CIDP patients during IVIg treatment, but further studies in larger cohorts are warranted to explore their utility to serve as a potential therapeutic biomarker for IVIg treatment response in CIDP.
Collapse
Affiliation(s)
- Christian Ritter
- Department of Neurology, University Hospital Cologne, Cologne, Germany. .,Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany. .,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Jülich, Jülich, Germany.
| | - Ilja Bobylev
- Department of Neurology, University Hospital Cologne, Cologne, Germany. .,Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Helmar C Lehmann
- Department of Neurology, University Hospital Cologne, Cologne, Germany. .,Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
12
|
IVIg for relapsing–remitting multiple sclerosis: promises and uncertainties. Trends Pharmacol Sci 2015; 36:419-21. [DOI: 10.1016/j.tips.2015.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 04/24/2015] [Indexed: 11/21/2022]
|
13
|
Tzekova N, Heinen A, Bunk S, Hermann C, Hartung HP, Reipert B, Küry P. Immunoglobulins stimulate cultured Schwann cell maturation and promote their potential to induce axonal outgrowth. J Neuroinflammation 2015; 12:107. [PMID: 26022648 PMCID: PMC4450464 DOI: 10.1186/s12974-015-0331-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/20/2015] [Indexed: 02/06/2023] Open
Abstract
Background Schwann cells are the myelinating glial cells of the peripheral nervous system and exert important regenerative functions revealing them as central repair components of many peripheral nerve pathologies. Intravenous immunoglobulins (IVIG) are widely used to treat autoimmune and inflammatory diseases including immune-mediated neuropathies. Nevertheless, promotion of peripheral nerve regeneration is currently an unmet therapeutical goal. We therefore examined whether immunoglobulins affect glial cell homeostasis, differentiation, and Schwann cell dependent nerve regenerative processes. Methods The responses of different primary Schwann cell culture models to IVIG were investigated: immature or differentiation competent Schwann cells, myelinating neuron/glial cocultures, and dorsal root ganglion explants. Immature or differentiating Schwann cells were used to study cellular proliferation, morphology, and gene/protein expression. Myelination rates were determined using myelinating neuron/glia cocultures, whereas axonal outgrowth was assessed using non-myelinating dorsal root ganglion explants. Results We found that IVIG specifically bind to Schwann cells and detected CD64 Fc receptor expression on their surface. In response to IVIG binding, Schwann cells reduced proliferation rates and accelerated growth of cellular protrusions. Furthermore, we observed that IVIG treatment transiently boosts myelin gene expression and myelination-related signaling pathways of immature cells, whereas in differentiating Schwann cells, myelin expression is enhanced on a long-term scale. Importantly, myelin gene upregulation was not detected upon application of IgG1 control antibodies. In addition, we demonstrate for the first time that Schwann cells secrete interleukin-18 upon IVIG stimulation and that this cytokine instructs these cells to promote axonal growth. Conclusions We conclude that IVIG can positively influence the Schwann cell differentiation process and that it enhances their regenerative potential.
Collapse
Affiliation(s)
- Nevena Tzekova
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - André Heinen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - Sebastian Bunk
- Department of Immunology, Baxter Innovations GmbH, Vienna, Austria.
| | - Corinna Hermann
- Medical Affairs EMEA, Baxter Innovations GmbH, Vienna, Austria.
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - Birgit Reipert
- Department of Immunology, Baxter Innovations GmbH, Vienna, Austria.
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
14
|
Klein D, Groh J, Weishaupt A, Martini R. Endogenous antibodies contribute to macrophage-mediated demyelination in a mouse model for CMT1B. J Neuroinflammation 2015; 12:49. [PMID: 25879857 PMCID: PMC4364634 DOI: 10.1186/s12974-015-0267-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/17/2015] [Indexed: 01/02/2023] Open
Abstract
Background We could previously identify components of both the innate and the adaptive immune system as disease modifiers in the pathogenesis of models for Charcot-Marie-Tooth (CMT) neuropathies type 1B and 1X. As part of the adaptive immune system, here we investigated the role of antibodies in a model for CMT1B. Methods Antibodies were localized and characterized in peripheral nerves of the CMT1B model by immunohistochemistry and Western blot analysis. Experimental ablation of antibodies was performed by cross breeding the CMT1B models with mutants deficient in B-lymphocytes (JHD−/− mutants). Ameliorated demyelination by antibody deficiency was reverted by intravenous injection of mouse IgG fractions. Histopathological analysis was performed by immunocytochemistry and light and quantitative electron microscopy. Results We demonstrate that in peripheral nerves of a mouse model for CMT1B, endogenous antibodies strongly decorate endoneurial tubes of peripheral nerves. These antibodies comprise IgG and IgM subtypes and are preferentially, but not exclusively, associated with nerve fiber aspects nearby the nodes of Ranvier. In the absence of antibodies, the early demyelinating phenotype is substantially ameliorated. Reverting the neuropathy by reconstitution with murine IgG fractions identified accumulating antibodies as potentially pathogenic at this early stage of disease. Conclusions Our study demonstrates that in a mouse model for CMT1B, endogenous antibodies contribute to early macrophage-mediated demyelination and disease progression. Thus, both the innate and adaptive immune system are mutually interconnected in a genetic model for demyelination. Since in Wallerian degeneration antibodies have also been shown to be involved in myelin phagocytosis, our study supports our view that inherited demyelination and Wallerian degeneration share common mechanisms, which are detrimental when activated under nonlesion conditions.
Collapse
Affiliation(s)
- Dennis Klein
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str 11, D-97080, Würzburg, Germany.
| | - Janos Groh
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str 11, D-97080, Würzburg, Germany.
| | - Andreas Weishaupt
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str 11, D-97080, Würzburg, Germany.
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Josef-Schneider-Str 11, D-97080, Würzburg, Germany.
| |
Collapse
|
15
|
IVIG regulates BAFF expression in patients with chronic inflammatory demyelinating polyneuropathy (CIDP). J Neuroimmunol 2014; 274:225-9. [PMID: 25002077 DOI: 10.1016/j.jneuroim.2014.06.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/10/2014] [Accepted: 06/12/2014] [Indexed: 12/24/2022]
Abstract
Recent studies indicate that the cytokine B-cell activating factor (BAFF) is involved in the pathogenesis of chronic inflammatory demyelinating polyneuropathy (CIDP). Intravenous immunoglobulin (IVIg) is standard treatment for CIDP and is known to rapidly modulate increased serum levels of pro-inflammatory cytokines. We evaluated the expression profile of BAFF and its corresponding BAFF-receptor in samples from CIDP patients, focusing on rapid changes before and after IVIg treatment. In CIDP patients BAFF serum concentrations were elevated compared to controls. Treatment with high-dose IVIg restored those elevated BAFF serum levels. Whereas treatment with IVIg did not affect BAFF production in monocytes, antibodies against BAFF could be detected in IVIg preparations, which may explain the short-term decrease of BAFF levels after IVIg treatment. Our data suggest that BAFF plays an important role in the pathogenesis of CIDP and may serve as marker for IVIg treatment response.
Collapse
|