1
|
Alipour R, Sereshki N, Rafiee M, Reza Mofid M, Alsahebfosoul F, Pourazar A. Blood IgMs from healthy donors and patients with systemic lupus erythematosus reduce the inflammatory properties of platelets from healthy donors. Immunobiology 2022; 227:152193. [DOI: 10.1016/j.imbio.2022.152193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 11/27/2022]
|
2
|
López P, Rodríguez-Carrio J, Martínez-Zapico A, Pérez-Álvarez ÁI, Benavente L, Caminal-Montero L, Suárez A. IgM anti-phosphorylcholine antibodies associate with senescent and IL-17+ T cells in SLE patients with a pro-inflammatory lipid profile. Rheumatology (Oxford) 2020; 59:407-417. [PMID: 31302689 DOI: 10.1093/rheumatology/kez264] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/04/2019] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE The aim was to evaluate whether T cell subsets and the lipid profile could be linked to the cardioprotective effect of IgM anti-phosphorylcholine (PC) antibodies in SLE. METHODS Anti-PC antibodies were quantified by ELISA in 197 patients and 99 controls and analysed in relationship to clinical features, treatments and serum lipids. Carotid atheromatosis was evaluated by ultrasonography; Th1, Th17, Treg and CD4+CD28null cells by flow cytometry; and cytokine serum levels by immunoassays, in a subgroup of 120 SLE patients and 33 controls. RESULTS IgM anti-PC serum levels were reduced in SLE patients compared with controls (P < 0.001) and were associated with age (β= -0.252; P = 0.002), high-density lipoprotein (HDL; β = 0.271; P = 0.001), low-density lipoprotein (LDL; β= -0.192; P = 0.017) and glucocorticoid treatment (β= -0.201; P = 0.012), whereas the IgG-to-IgM anti-PC ratio was increased (P = 0.007) and associated with age (β = 0.194; P = 0.028) and SLEDAI (β = 0.250; P = 0.005). Also, patients with clinical or subclinical cardiovascular disease exhibited reduced IgM anti-PC levels compared with their cardiovascular disease-free counterparts, regardless of glucocorticoid usage (P = 0.001). CD4+CD28null and Th17 cells were increased in SLE patients compared with controls (P < 0.01) and correlated inversely with IgM anti-PC levels. These associations were observed in patients displaying high triglyceride or low HDL levels, even after adjusting for clinical parameters and treatments (CD4+CD28null: β = -0.455, P = 0.001; Th17: β= -0.280, P = 0.035), but not in those with a normal lipid profile. High triglyceride and low HDL profiles were related to low IgM anti-PC and Treg levels, respectively, whereas both lipid profiles were associated with inflammatory markers and cytokines. CONCLUSION The present study provides evidence for an association of IgM anti-PC antibodies with pro-atherogenic T cell subsets in SLE, with a high triglyceride/low HDL lipid profile playing a facilitating major role.
Collapse
Affiliation(s)
- Patricia López
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Aleida Martínez-Zapico
- Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Lorena Benavente
- Department of Neurology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Luis Caminal-Montero
- Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA).,Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana Suárez
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| |
Collapse
|
3
|
Lobo PI, Okusa MD. Role of Natural IgM and IgM Induced Bregs in Preventing Ischemia Induced Innate Inflammation and Acute Kidney Injury. Nephron Clin Pract 2019; 143:166-169. [PMID: 31288230 DOI: 10.1159/000501639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/20/2019] [Indexed: 11/19/2022] Open
Abstract
Ischemic acute kidney injury (AKI) is predominantly mediated by the innate inflammatory response to damage-associated molecular patterns released during the reperfusion phase of the ischemic event. In this study, we show that pre-emptive IgM infusions to increase binding of natural IgM (nIgM) anti-leucocyte autoantibodies (IgM-ALA), inhibit this inflammatory response and prevent AKI in mice. Similarly, AKI was prevented by pre-emptively infusing Bregs, induced ex vivo by pre-treating pan-B cells with nIgM. Harnessing such a physiologic mechanism to inhibit inflammation and prevent ischemia-induced AKI can have translational potential in humans. For example, one can pre-emptively infuse IgM or ex vivo induced Bregs in patients who have a high risk of developing ischemia-induced AKI, especially the subset of these patients with low levels of IgM-ALA or regulatory T cells (Tregs).
Collapse
Affiliation(s)
- Peter I Lobo
- Division of Nephrology and CIIR, University of Virginia Health System, Charlottesville, Virginia, USA,
| | - Mark D Okusa
- Division of Nephrology and CIIR, University of Virginia Health System, Charlottesville, Virginia, USA
| |
Collapse
|
4
|
Platt JL, Cascalho M. Non-canonical B cell functions in transplantation. Hum Immunol 2019; 80:363-377. [PMID: 30980861 PMCID: PMC6544480 DOI: 10.1016/j.humimm.2019.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022]
Abstract
B cells are differentiated to recognize antigen and respond by producing antibodies. These activities, governed by recognition of ancillary signals, defend the individual against microorganisms and the products of microorganisms and constitute the canonical function of B cells. Despite the unique differentiation (e.g. recombination and mutation of immunoglobulin gene segments) toward this canonical function, B cells can provide other, "non-canonical" functions, such as facilitating of lymphoid organogenesis and remodeling and fashioning T cell repertoires and modifying T cell responses. Some non-canonical functions are exerted by antibodies, but most are mediated by other products and/or direct actions of B cells. The diverse set of non-canonical functions makes the B cell as much as any cell a central organizer of innate and adaptive immunity. However, the diverse products and actions also confound efforts to weigh the importance of individual non-canonical B cell functions. Here we shall describe the non-canonical functions of B cells and offer our perspective on how those functions converge in the development and governance of immunity, particularly immunity to transplants, and hurdles to advancing understanding of B cell functions in transplantation.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Departments of Surgery and of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States.
| | - Marilia Cascalho
- Departments of Surgery and of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
5
|
Guo Z, Tao Y, Yin S, Song Y, Lu X, Li X, Fan Y, Fan X, Xu S, Yang J, Yu Y. The transcription factor Foxp1 regulates the differentiation and function of dendritic cells. Mech Dev 2019; 158:103554. [PMID: 31077741 DOI: 10.1016/j.mod.2019.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 01/14/2023]
Abstract
Dendritic cells (DCs) are the sentinels of the immune system and play a critical role in initiating adaptive immune responses against pathogens. As the most powerful antigen presenting cells, DCs are also important in maintaining immune homeostasis and participating in the development of autoimmune diseases. How the maturation and function of DCs is regulated in these conditions and what is the function of various transcription factors is still unclear. In this study, we found that the expression of the transcription factor Foxp1 gradually increased during the maturation of DCs. Then, we constructed a recombinant adenovirus carrying Foxp1-interfering RNA (Ad-simFoxp1) and transfected murine bone marrow-derived DCs in vitro. DCs transfected with Ad-simFoxp1 exhibited markedly lower costimulatory molecules, and decreased cytokines. And Ad-simFoxp1 greatly inhibited mature DC-induced T cell responses. Moreover, in vivo infusion with Ad-simFoxp1-modified DCs significantly delayed the onset of experimental autoimmune encephalomyelitis (EAE). Therefore, adoptive transfection of Ad-simFoxp1 in DCs may be a potential treatment strategy against autoimmune diseases.
Collapse
Affiliation(s)
- Ziyi Guo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China; Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Yijie Tao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Shulei Yin
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yuping Song
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaomin Lu
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Xuesong Li
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Yujuan Fan
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaofang Fan
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Sheng Xu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China.
| | - Jialin Yang
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yizhi Yu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China.
| |
Collapse
|
6
|
Yang Y, Chen L, Sun HW, Guo H, Song Z, You Y, Yang LY, Tong YN, Gao JN, Zeng H, Yang WC, Zou QM. Epitope-loaded nanoemulsion delivery system with ability of extending antigen release elicits potent Th1 response for intranasal vaccine against Helicobacter pylori. J Nanobiotechnology 2019; 17:6. [PMID: 30660182 PMCID: PMC6339695 DOI: 10.1186/s12951-019-0441-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/03/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection remains a global public health issue, especially in Asia. Due to the emergence of antibiotic-resistant strains and the complexity of H. pylori infection, conventional vaccination is the best way to control the disease. Our previous study found that the N-acetyl-neuroaminyllactose-binding hemagglutinin protein (HpaA) is an effective protective antigen for vaccination against H. pylori infection, and intranasal immunization with the immunodominant HpaA epitope peptide (HpaA 154-171, P22, MEGVLIPAGFIKVTILEP) in conjunction with a CpG adjuvant decreased bacterial colonization in H. pylori-infected mice. However, to confer more robust and effective protection against H. pylori infection, an optimized delivery system is needed to enhance the P22-specific memory T cell response. RESULTS In this study, an intranasal nanoemulsion (NE) delivery system offering high vaccine efficacy without obvious cytotoxicity was designed and produced. We found that this highly stable system significantly prolonged the nasal residence time and enhanced the cellular uptake of the epitope peptide, which powerfully boosted the specific Th1 responses of the NE-P22 vaccine, thus reducing bacterial colonization without CpG. Furthermore, the protection efficacy was further enhanced by combining the NE-P22 vaccine with CpG. CONCLUSION This epitope-loaded nanoemulsion delivery system was shown to extend antigen release and elicit potent Th1 response, it is an applicable delivery system for intranasal vaccine against H. pylori.
Collapse
Affiliation(s)
- Yun Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Li Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Hong-wu Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hong Guo
- Department of Gastroenterology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Zhen Song
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ying You
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Liu-yang Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ya-nan Tong
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ji-ning Gao
- Institute of Combined Injury of PLA, College of Military Preventive Medicine, Third Military Medical University of Chinese PLA, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wu-chen Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
- Department of Hematology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Quan-ming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
7
|
Fereidan-Esfahani M, Nayfeh T, Warrington A, Howe CL, Rodriguez M. IgM Natural Autoantibodies in Physiology and the Treatment of Disease. Methods Mol Biol 2019; 1904:53-81. [PMID: 30539466 DOI: 10.1007/978-1-4939-8958-4_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibodies are vital components of the adaptive immune system for the recognition and response to foreign antigens. However, some antibodies recognize self-antigens in healthy individuals. These autoreactive antibodies may modulate innate immune functions. IgM natural autoantibodies (IgM-NAAs) are a class of primarily polyreactive immunoglobulins encoded by germline V-gene segments which exhibit low affinity but broad specificity to both foreign and self-antigens. Historically, these autoantibodies were closely associated with autoimmune disease. Nevertheless, not all human autoantibodies are pathogenic and compelling evidence indicates that IgM-NAAs may exert a spectrum of effects from injurious to protective depending upon cellular and molecular context. In this chapter, we review the current state of knowledge regarding the potential physiological and therapeutic roles of IgM-NAAs in different disease conditions such as atherosclerosis, cancer, and autoimmune disease. We also describe the discovery of two reparative IgM-NAAs by our laboratory and delineate their proposed mechanisms of action in central nervous system (CNS) disease.
Collapse
Affiliation(s)
| | - Tarek Nayfeh
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
| | | |
Collapse
|
8
|
Chhabra P, Spano AJ, Bowers D, Ren T, Moore DJ, Timko MP, Wu M, Brayman KL. Evidence for the Role of the Cecal Microbiome in Maintenance of Immune Regulation and Homeostasis. Ann Surg 2018; 268:541-549. [PMID: 29994931 DOI: 10.1097/sla.0000000000002930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE (S) Our objective was to investigate alterations in the cecal microbial composition during the development of type 1 diabetes (T1D) with or without IgM therapy, and correlate these alterations with the corresponding immune profile. METHODS (1) Female nonobese diabetic (NOD) mice treated with IgM or saline (n = 20/group) were divided into 5-week-old nondiabetic; 9 to 12-week-old prehyperglycemic stage-1; ≥13-week-old prehyperglycemic stage-2; and diabetic groups. 16S rRNA libraries were prepared from bacterial DNA and deep-sequenced. (2) New-onset diabetic mice were treated with IgM (200 μg on Days 1, 3, and 5) and their blood glucose monitored for 2 months. RESULTS Significant dysbiosis was observed in the cecal microbiome with the progression of T1D development. The alteration in microbiome composition was characterized by an increase in the bacteroidetes:firmicutes ratio. In contrast, IgM conserved normal bacteroidetes:firmicutes ratio and this effect was long-lasting. Furthermore, oral gavage using cecal content from IgM-treated mice significantly diminished the incidence of diabetes compared with controls, indicating that IgM specifically affected mucosa-associated microbes, and that the affect was causal and not an epiphenomenon. Also, regulatory immune cell populations (myeloid-derived suppressor cells and regulatory T cells) were expanded and insulin autoantibody production diminished in the IgM-treated mice. In addition, IgM therapy reversed hyperglycemia in 70% of new-onset diabetic mice (n = 10) and the mice remained normoglycemic for the entire post-treatment observation period. CONCLUSIONS The cecal microbiome appears to be important in maintaining immune homeostasis and normal immune responses.
Collapse
Affiliation(s)
- Preeti Chhabra
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Anthony J Spano
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Daniel Bowers
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Tiantian Ren
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Daniel J Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN
| | - Michael P Timko
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Martin Wu
- Department of Biology, University of Virginia, Charlottesville, VA
| | | |
Collapse
|
9
|
Song N, Zhang T, Xu X, Lu Z, Yu X, Fang Y, Hu J, Jia P, Teng J, Ding X. miR-21 Protects Against Ischemia/Reperfusion-Induced Acute Kidney Injury by Preventing Epithelial Cell Apoptosis and Inhibiting Dendritic Cell Maturation. Front Physiol 2018; 9:790. [PMID: 30013485 PMCID: PMC6036242 DOI: 10.3389/fphys.2018.00790] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/06/2018] [Indexed: 02/02/2023] Open
Abstract
Renal tubular injury and innate immune responses induced by hypoxia contribute to acute kidney injury. Accumulating evidence suggests that miR-21 overexpression protects against kidney ischemia injury. Additionally, miR-21 emerges as a key inhibitor in dendritic cell maturation. Thus, we hypothesized that miR-21 protects the kidney from IR injury by suppressing epithelial cell damage and inflammatory reaction. In this study, we investigated effects of miR-21 and its signaling pathways (PTEN/AKT/mTOR/HIF, PDCD4/NFκ-B) on kidney ischemia/reperfusion (IR) injury in vitro and in vivo. The results revealed that IR increased miR-21, HIF1α, and 2α expression in vivo and in vitro. MiR-21 interacted with HIF1α and 2α through the PTEN/AKT/mTOR pathway. Moreover, inhibition of miR-21 activated PDCD4/NFκ-B pathways, which are critical for dendritic cell maturation. Renal IR triggers local inflammation by inducing the dendritic cell maturation and promoting the secretion of IL-12, IL-6, and TNF-α cytokines. Knockdown of miR-21 intensified the effect of IR on tubular epithelial cell apoptosis and dendritic cell maturation. Our results suggested that IR-inducible miR-21 protects epithelial cells from IR injury via a feedback interaction with HIF (PTEN/AKT/mTOR/HIF/miR-21) and by inhibiting maturation of DCs through the PDCD4/NF-κB pathway. These findings highlight new therapeutic opportunities in AKI.
Collapse
Affiliation(s)
- Nana Song
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Ting Zhang
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - XiaLian Xu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Zhihui Lu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaofang Yu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Yi Fang
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jiachang Hu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Ping Jia
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jie Teng
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| |
Collapse
|
10
|
Luo XQ, Shao JB, Xie RD, Zeng L, Li XX, Qiu SQ, Geng XR, Yang LT, Li LJ, Liu DB, Liu ZG, Yang PC. Micro RNA-19a interferes with IL-10 expression in peripheral dendritic cells of patients with nasal polyposis. Oncotarget 2018; 8:48915-48921. [PMID: 28388587 PMCID: PMC5564735 DOI: 10.18632/oncotarget.16555] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/14/2017] [Indexed: 01/01/2023] Open
Abstract
The pathogenesis of nasal polyp is to be further investigated. Micro RNA (miR) plays a role in the development of allergic inflammation. Interleukin (IL)-10-producing dendritic cells (DC) have immune tolerogenic properties. This study test a hypothesis that miR-17-92 cluster is associated with suppressing IL-10 in peripheral DC. In this study, peripheral blood samples were obtained from 26 patients with nasal polyp. The CD11c DCs were isolated from the blood samples and analyzed for the expression of IL-10. We observed that, as compared with healthy subjects, the IL-10 expression in peripheral DC was significantly lower in polyp patients. The levels of miR-19a, but not the rest 5 members of the miR-17-92 cluster, were markedly higher in DCs in polyp group. Exposure to recombinant IL-4 suppressed the IL-10 expression in DCs, which was abolished by blocking histone deacetylase-11 or knocking down the miR-19a gene in DCs. We conclude that miR-19a plays a critical role in the suppression of IL-10 in peripheral DCs, which may be a target in the immune therapy for nasal polyp.
Collapse
Affiliation(s)
- Xiang-Qian Luo
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510010, China.,The Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Jian-Bo Shao
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510010, China.,The Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Rui-Di Xie
- The Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Lu Zeng
- The Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Xiao-Xi Li
- The Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Shu-Qi Qiu
- Longgang ENT Hospital, Shenzhen 518116, China
| | | | - Li-Tao Yang
- Longgang ENT Hospital, Shenzhen 518116, China.,Brain Body Institute, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Lin-Jing Li
- Brain Body Institute, McMaster University, Hamilton, ON, L8N 4A6, Canada
| | - Da-Bo Liu
- Department of Otolaryngology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510010, China
| | - Zhi-Gang Liu
- The Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Ping-Chang Yang
- The Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| |
Collapse
|
11
|
IgM antibodies against phosphorylcholine promote polarization of T regulatory cells from patients with atherosclerotic plaques, systemic lupus erythematosus and healthy donors. Atherosclerosis 2018; 268:36-48. [DOI: 10.1016/j.atherosclerosis.2017.11.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 10/18/2017] [Accepted: 11/15/2017] [Indexed: 12/20/2022]
|
12
|
Bello-Gil D, Khasbiullina N, Shilova N, Bovin N, Mañez R. Repertoire of BALB/c Mice Natural Anti-Carbohydrate Antibodies: Mice vs. Humans Difference, and Otherness of Individual Animals. Front Immunol 2017; 8:1449. [PMID: 29163519 PMCID: PMC5681490 DOI: 10.3389/fimmu.2017.01449] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/17/2017] [Indexed: 01/12/2023] Open
Abstract
One of the most common genetic backgrounds for mice used as a model to investigate human diseases is the inbred BALB/c strain. This work is aimed to characterize the pattern of natural anti-carbohydrate antibodies present in the serum of 20 BALB/c mice by printed glycan array technology and to compare their binding specificities with that of human natural anti-carbohydrate antibodies. Natural antibodies (NAbs) from the serum of BALB/c mice interacted with 71 glycans from a library of 419 different carbohydrate structures. However, only seven of these glycans were recognized by the serum of all the animals studied, and other five glycans by at least 80% of mice. The pattern of the 12 glycans mostly recognized by the circulating antibodies of BALB/c mice differed significantly from that observed with natural anti-carbohydrate antibodies in humans. This lack of identical repertoires of natural anti-carbohydrate antibodies between individual inbred mice, and between mice and humans, should be taken into consideration when mouse models are intended to be used for investigation of NAbs in biomedical research.
Collapse
Affiliation(s)
- Daniel Bello-Gil
- Infectious Pathology and Transplantation Division, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Nailya Khasbiullina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda Shilova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nicolai Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Rafael Mañez
- Infectious Pathology and Transplantation Division, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Intensive Care Department, Bellvitge University Hospital, Hospitalet de Llobregat, Spain
| |
Collapse
|
13
|
Lobo PI, Schlegel KH, Bajwa A, Huang L, Okusa MD. Natural IgM and TLR Agonists Switch Murine Splenic Pan-B to "Regulatory" Cells That Suppress Ischemia-Induced Innate Inflammation via Regulating NKT-1 Cells. Front Immunol 2017; 8:974. [PMID: 28878768 PMCID: PMC5572342 DOI: 10.3389/fimmu.2017.00974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/31/2017] [Indexed: 01/01/2023] Open
Abstract
Natural IgM anti-leukocyte autoantibodies (IgM-ALAs) inhibit inflammation by several mechanisms. Here, we show that pan-B cells and bone marrow-derived dendritic cells (BMDCs) are switched to regulatory cells when pretreated ex vivo with IgM. B cells are also switched to regulatory cells when pretreated ex vivo with CpG but not with LPS. Pre-emptive infusion of such ex vivo induced regulatory cells protects C57BL/6 mice from ischemia-induced acute kidney injury (AKI) via regulation of in vivo NKT-1 cells, which normally amplify the innate inflammatory response to DAMPS released after reperfusion of the ischemic kidney. Such ex vivo induced regulatory pan-B cells and BMDC express low CD1d and inhibit inflammation by regulating in vivo NKT-1 in the context of low-lipid antigen presentation and by a mechanism that requires costimulatory molecules, CD1d, PDL1/PD1, and IL10. Second, LPS and CpG have opposite effects on induction of regulatory activity in BMDC and B cells. LPS enhances regulatory activity of IgM-pretreated BMDC but negates the IgM-induced regulatory activity in B cells, while CpG, with or without IgM pretreatment, induces regulatory activity in B cells but not in BMDC. Differences in the response of pan-B and dendritic cells to LPS and CpG, especially in the presence of IgM-ALA, may have relevance during infections and inflammatory disorders where there is an increased IgM-ALA and release of TLRs 4 and 9 ligands. Ex vivo induced regulatory pan-B cells could have therapeutic relevance as these easily available cells can be pre-emptively infused to prevent AKI that can occur during open heart surgery or in transplant recipients receiving deceased donor organs.
Collapse
Affiliation(s)
- Peter I Lobo
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA, United States
| | - Kailo H Schlegel
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA, United States
| | - Amandeep Bajwa
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA, United States
| | - Liping Huang
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA, United States
| | - Mark D Okusa
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
14
|
Wu X, Dayanand KK, Thylur RP, Norbury CC, Gowda DC. Small molecule-based inhibition of MEK1/2 proteins dampens inflammatory responses to malaria, reduces parasite load, and mitigates pathogenic outcomes. J Biol Chem 2017; 292:13615-13634. [PMID: 28679535 DOI: 10.1074/jbc.m116.770313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 06/23/2017] [Indexed: 12/16/2022] Open
Abstract
Malaria infections cause several systemic and severe single- or multi-organ pathologies, killing hundreds of thousands of people annually. Considering the existing widespread resistance of malaria parasites to anti-parasitic drugs and their high propensity to develop drug resistance, alternative strategies are required to manage malaria infections. Because malaria is a host immune response-driven disease, one approach is based on gaining a detailed understanding of the molecular and cellular processes that modulate malaria-induced innate and adaptive immune responses. Here, using a mouse cerebral malaria model and small-molecule inhibitors, we demonstrate that inhibiting MEK1/2, the upstream kinases of ERK1/2 signaling, alters multifactorial components of the innate and adaptive immune responses, controls parasitemia, and blocks pathogenesis. Specifically, MEK1/2 inhibitor treatment up-regulated B1 cell expansion, IgM production, phagocytic receptor expression, and phagocytic activity, enhancing parasite clearance by macrophages and neutrophils. Further, the MEK1/2 inhibitor treatment down-regulated pathogenic pro-inflammatory and helper T cell 1 (Th1) responses and up-regulated beneficial anti-inflammatory cytokine responses and Th2 responses. These inhibitor effects resulted in reduced granzyme B expression by T cells, chemokine and intracellular cell adhesion molecule 1 (ICAM-1) expression in the brain, and chemokine receptor expression by both myeloid and T cells. These bimodal effects of the MEK1/2 inhibitor treatment on immune responses contributed to decreased parasite biomass, organ inflammation, and immune cell recruitment, preventing tissue damage and death. In summary, we have identified several previously unrecognized immune regulatory processes through which a MEK1/2 inhibitor approach controls malaria parasitemia and mitigates pathogenic effects on host organs.
Collapse
Affiliation(s)
- Xianzhu Wu
- From the Departments of Biochemistry and Molecular Biology and
| | | | - Ramesh P Thylur
- From the Departments of Biochemistry and Molecular Biology and
| | - Christopher C Norbury
- Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - D Channe Gowda
- From the Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
15
|
Lobo PI. Role of Natural IgM Autoantibodies (IgM-NAA) and IgM Anti-Leukocyte Antibodies (IgM-ALA) in Regulating Inflammation. Curr Top Microbiol Immunol 2017; 408:89-117. [PMID: 28698955 DOI: 10.1007/82_2017_37] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Natural IgM autoantibodies (IgM-NAA) are rapidly produced to inhibit pathogens and abrogate inflammation mediated by invading microorganisms and host neoantigens. IgM-NAA achieve this difficult task by being polyreactive with low binding affinity but with high avidity, characteristics that allow these antibodies to bind antigenic determinants shared by pathogens and neoantigens. Hence the same clones of natural IgM can bind and mask host neoantigens as well as inhibit microorganisms. In addition, IgM-NAA regulate the inflammatory response via mechanisms involving binding of IgM to apoptotic cells to enhance their removal and binding of IgM to live leukocytes to regulate their function. Secondly, we review how natural IgM prevents autoimmune disorders arising from pathogenic IgG autoantibodies as well as by autoreactive B and T cells that have escaped tolerance mechanisms. Thirdly, using IgM knockout mice, we show that regulatory B and T cells require IgM to effectively regulate inflammation mediated by innate, adaptive and autoimmune mechanisms. It is therefore not surprising why the host positively selects such autoreactive B1 cells that generate protective IgM-NAA, which are also evolutionarily conserved. Fourthly, we show that IgM anti-leukocyte autoantibodies (IgM-ALA) levels and their repertoire can vary in normal humans and disease states and this variation may partly explain the observed differences in the inflammatory response after infection, ischemic injury or after a transplant. Finally we also show how protective IgM-NAA can be rendered pathogenic under non-physiological conditions. IgM-NAA have therapeutic potential. Polyclonal IgM infusions can be used to abrogate ongoing inflammation. Additionally, inflammation arising after ischemic kidney injury, e.g., during high-risk elective cardiac surgery or after allograft transplantation, can be prevented by pre-emptively infusing polyclonal IgM, or DC pretreated ex vivo with IgM, or by increasing in vivo IgM with a vaccine approach. Cell therapy with IgM pretreated cells, is appealing as less IgM will be required.
Collapse
Affiliation(s)
- Peter I Lobo
- Department of Internal Medicine, Division of Nephrology, Center of Immunology, Inflammation and Regenerative Medicine, University of Virginia Health Center, Charlottesville, VA, USA.
| |
Collapse
|
16
|
Lobo PI. Role of Natural Autoantibodies and Natural IgM Anti-Leucocyte Autoantibodies in Health and Disease. Front Immunol 2016; 7:198. [PMID: 27375614 PMCID: PMC4893492 DOI: 10.3389/fimmu.2016.00198] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/06/2016] [Indexed: 11/13/2022] Open
Abstract
We review how polyreactive natural IgM autoantibodies (IgM-NAA) protect the host from invading micro-organisms and host neo-antigens that are constantly being produced by oxidation mechanisms and cell apoptosis. Second, we discuss how IgM-NAA and IgM anti-leukocyte antibodies (IgM-ALA) inhibits autoimmune inflammation by anti-idiotypic mechanisms, enhancing removal of apoptotic cells, masking neo-antigens, and regulating the function of dendritic cells (DC) and effector cells. Third, we review how natural IgM prevents autoimmune disorders arising from pathogenic IgG autoantibodies, triggered by genetic mechanisms (e.g., SLE) or micro-organisms, as well as by autoreactive B and T cells that have escaped tolerance mechanisms. Studies in IgM knockout mice have clearly demonstrated that regulatory B and T cells require IgM to effectively regulate inflammation mediated by innate, adaptive, and autoimmune mechanisms. It is, therefore, not surprising why the host positively selects such autoreactive B1 cells that generate IgM-NAA, which are also evolutionarily conserved. Fourth, we show that IgM-ALA levels and their repertoire can vary in normal humans and disease states and this variation may partly explain the observed differences in the inflammatory response after infection, ischemic injury, or after a transplant. We also show how protective IgM-NAA can be rendered pathogenic under non-physiological conditions. We also review IgG-NAA that are more abundant than IgM-NAA in plasma. However, we need to understand if the (Fab)(2) region of IgG-NAA has physiological relevance in non-disease states, as in plasma, their functional activity is blocked by IgM-NAA having anti-idiotypic activity. Some IgG-NAA are produced by B2 cells that have escaped tolerance mechanisms and we show how such pathogenic IgG-NAA are regulated to prevent autoimmune disease. The Fc region of IgG-NAA can influence inflammation and B cell function in vivo by binding to activating and inhibitory FcγR. IgM-NAA has therapeutic potential. Polyclonal IgM infusions can be used to abrogate on-going inflammation. Additionally, inflammation arising after ischemic kidney injury, e.g., during high-risk elective cardiac surgery or after allograft transplantation, can be prevented by pre-emptively infusing polyclonal IgM or DC pretreated ex vivo with IgM or by increasing in vivo IgM with a vaccine approach. Cell therapy is appealing as less IgM will be required.
Collapse
Affiliation(s)
- Peter Isaac Lobo
- Department of Internal Medicine, Division of Nephrology, Center of Immunology, Inflammation and Regenerative Medicine, University of Virginia Health Center, Charlottesville, VA, USA
| |
Collapse
|