1
|
Zipfel PF, Heidenreich K. The 4 functional segments of Factor H: Role in physiological target recognition and contribution to disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf065. [PMID: 40356067 DOI: 10.1093/jimmun/vkaf065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/07/2025] [Indexed: 05/15/2025]
Abstract
Factor H controls proximal complement activation, and its dysfunction leads to diseases that often manifest in the kidney. Structural and functional analyses have identified 4 distinct functional segments: an N-terminal regulatory unit, a cell binding unit, a segment with combined low-affinity C3b and heparin sites, and a C-terminal recognition or sensor unit with overlapping C3b/C3d and heparin sites. Three segments are linked to diseases. The regulatory segment is affected in C3 glomerulopathy and antineutrophil cytoplasmic antibody-associated vasculitis. The second segment includes the Y402H polymorphism of age-related macular degeneration, is associated with different types of cancer, and is targeted by pathogens. The C-terminal sensor segment is involved in atypical hemolytic uremic syndrome, in FHR1:FHR3 deficient and autoantibody-positive hemolytic uremic syndrome form and is exploited by pathogens. Factor H function is modulated by Factor H like protein 1 and FHR1, 2 plasma proteins that share segments with Factor H. This interplay is critical for fine-tuning local complement. Understanding Factor H's physiological role, as well as the impact of its absence, mutations, or autoantibody targeting, provides insights into disease mechanisms and provides opportunities for therapeutic intervention by using full-length Factor H, its fragments, or complement-modulatory compounds.
Collapse
Affiliation(s)
- Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | | |
Collapse
|
2
|
Jeon C, Kim D, Kim KM, Lee SH, Lee JH, Kim SH, Kim JS, Kang YM, Jo S, Kim TH, Son CN. Complement factor H-related protein 5 alleviates joint inflammation and osteoclast differentiation by disrupting RANK-JNK signaling in collagen antibody-induced arthritis mouse model. Cytokine 2024; 184:156790. [PMID: 39461285 DOI: 10.1016/j.cyto.2024.156790] [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: 07/15/2024] [Revised: 10/07/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Complement Factor H-Related protein 5 (CFHR5) belongs to the factor H/CFHR family and regulates the complement system by modulating factor H's inhibitory activity against C3b. Despite its known role, the impact of CFHR5 on autoimmune arthritis and its relationship to pathophysiological changes in arthritis and bone loss remain unclear. This study aimed to assess the effect of CFHR5 on aggressive osteoclast activity and arthritis using a murine model of collagen antibody-induced arthritis (CAIA). METHODS The effect of recombinant CFHR5 protein (rCFHR5) on arthritis were evaluated in CAIA. The mice were divided into three group and intraperitoneally treated with rCFHR5, methotrexate (MTX) as positive control or PBS as negative control. In the CAIA mouse model, the rCFHR5-treated group significantly reduced the incidence and clinical arthritis equivalent to the MTX group. Clinical arthritis scores, incidence and body weight were measured, and histological analysis of ankle joints was performed by Hematoxylin and Eosin (H&E) and Safranin O - Fast green (SOFG), Tartrate-resistant acid phosphatase (TRAP) staining and Immunohistochemistry. Moreover, to investigate the rCFHR5 role, we isolated murine osteoclast precursor cells (OCPs) from each group, induced osteoclasts with M-CSF and RANKL, and performed TRAP and F-actin staining. To verify the mechanism, mRNA and protein analyses were performed in OCPs. RESULTS Histological examination of ankle joints revealed substantial reductions in synovial hyperplasia, bone marrow inflammation, bone erosion, cartilage destruction and TRAP-positive cells in the rCFHR5 group compared to the vehicle group. The ankle joints of the rCFHR5 group showed markedly decreased expression of proinflammatory cytokines (TNF-α, IL-1β and IL-6). Mechanically, treatment with rCFHR5 inhibited RANKL-mediated osteoclast differentiation from OCPs and disrupted the RANK-JNK signaling. These findings demonstrate that treatment with rCFHR5 attenuates joint inflammation and reduces osteoclast differentiation, indicating its potential anti-inflammatory effect in autoimmune arthritis models.
Collapse
Affiliation(s)
- Chanhyeok Jeon
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea; Deparment of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Dongju Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea; Deparment of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Kyung-Me Kim
- Department of Rheumatology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea; Eulji Rheumatology Research Institute, Eulji University, Uijeongbu, Republic of Korea
| | - Seung Hoon Lee
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea
| | - Ji-Hyun Lee
- Department of Rheumatology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea; Eulji Rheumatology Research Institute, Eulji University, Uijeongbu, Republic of Korea
| | - Sang-Hyon Kim
- Division of Rheumatology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jong-Seo Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young Mo Kang
- Preclina Inc, Incheon, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sungsin Jo
- Department of Biology, Soonchunhyang University, Asan, Republic of Korea.
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Republic of Korea; Deparment of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea; Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Republic of Korea.
| | - Chang-Nam Son
- Department of Rheumatology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea; Eulji Rheumatology Research Institute, Eulji University, Uijeongbu, Republic of Korea.
| |
Collapse
|
3
|
Zhao J, Zhang K, Sui D, Wang S, Li Y, Tang X, Liu X, Song Y, Deng Y. Recent advances in sialic acid-based active targeting chemoimmunotherapy promoting tumor shedding: a systematic review. NANOSCALE 2024; 16:14621-14639. [PMID: 39023195 DOI: 10.1039/d4nr01740d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Tumors have always been a major public health concern worldwide, and attempts to look for effective treatments have never ceased. Sialic acid is known to be a crucial element for tumor development and its receptors are highly expressed on tumor-associated immune cells, which perform significant roles in establishing the immunosuppressive tumor microenvironment and further boosting tumorigenesis, progression, and metastasis. Obviously, it is essential to consider sophisticated crosstalk between tumors, the immune system, and preparations, and understand the links between pharmaceutics and immunology. Sialic acid-based chemoimmunotherapy enables active targeting drug delivery via mediating the recognition between the sialic acid-modified nano-drug delivery system represented by liposomes and sialic acid-binding receptors on tumor-associated immune cells, which inhibit their activity and utilize their homing ability to deliver drugs. Such a "Trojan horse" strategy has remarkably improved the shortcomings of traditional passive targeting treatments, unexpectedly promoted tumor shedding, and persistently induced robust immunological memory, thus highlighting its prospective application potential for targeting various tumors. Herein, we review recent advances in sialic acid-based active targeting chemoimmunotherapy to promote tumor shedding, summarize the current viewpoints on the tumor shedding mechanism, especially the formation of durable immunological memory, and analyze the challenges and opportunities of this attractive approach.
Collapse
Affiliation(s)
- Jingyi Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Kunfeng Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yantong Li
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xueying Tang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| |
Collapse
|
4
|
Madden B, Singh RD, Haas M, Palma LMP, Sharma A, Vargas MJ, Gross L, Negron V, Nate T, Charlesworth MC, Theis JD, Nasr SH, Nath KA, Fervenza FC, Sethi S. Apolipoprotein E is enriched in dense deposits and is a marker for dense deposit disease in C3 glomerulopathy. Kidney Int 2024; 105:1077-1087. [PMID: 38447879 DOI: 10.1016/j.kint.2024.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/08/2024]
Abstract
C3 glomerulopathy (C3G) is a rare disease resulting from dysregulation of the alternative pathway of complement. C3G includes C3 glomerulonephritis (C3GN) and dense deposit disease (DDD), both of which are characterized by bright glomerular C3 staining on immunofluorescence studies. However, on electron microscopy (EM), DDD is characterized by dense osmiophilic mesangial and intramembranous deposits along the glomerular basement membranes (GBM), while the deposits of C3GN are not dense. Why the deposits appear dense in DDD and not in C3GN is not known. We performed laser microdissection (LCM) of glomeruli followed by mass spectrometry (MS) in 12 cases each of DDD, C3GN, and pretransplant kidney control biopsies. LCM/MS showed marked accumulation of complement proteins C3, C5, C6, C7, C8, C9 and complement regulating proteins CFHR5, CFHR1, and CFH in C3GN and DDD compared to controls. C3, CFH and CFHR proteins were comparable in C3GN and DDD. Yet, there were significant differences. First, there was a six-to-nine-fold increase of C5-9 in DDD compared to C3GN. Secondly, an unexpected finding was a nine-fold increase in apolipoprotein E (ApoE) in DDD compared to C3GN. Most importantly, immunohistochemical and confocal staining for ApoE mirrored the dense deposit staining in the GBM in DDD but not in C3GN or control cases. Validation studies using 31 C3G cases confirmed the diagnosis of C3GN and DDD in 80.6 % based on ApoE staining. Overall, there is a higher burden of terminal complement pathway proteins in DDD compared to C3GN. Thus, our study shows that dense deposits in DDD are enriched with ApoE compared to C3GN and control cases. Hence, ApoE staining may be used as an adjunct to EM for the diagnosis of DDD and might be valuable when EM is not available.
Collapse
Affiliation(s)
- Benjamin Madden
- Mayo Clinic Proteomics Core, Mayo Clinic, Rochester, Minnesota, USA
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark Haas
- Department of Pathology & Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lilian M P Palma
- Pediatric Nephrology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Alok Sharma
- Department of Renal Pathology & Electron Microscopy, Dr Lal Path Labs, New Delhi, India
| | - Maria J Vargas
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - LouAnn Gross
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vivian Negron
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Torell Nate
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jason D Theis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samih H Nasr
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Karl A Nath
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Fernando C Fervenza
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sanjeev Sethi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
5
|
Li J, Dong Y, Chen F, Yang H, Chen P, Li H, Shi S, Zhou X, Zhu L, Zhang Y, Liu L, Xie X, Yu F, Jin J, Lv J, Zhang H. Heterozygous mutations in factor H aggravate pathological damage in a stable IgA deposition model induced by Lactobacillus casei cell wall extract. Front Immunol 2024; 15:1368322. [PMID: 38558821 PMCID: PMC10978756 DOI: 10.3389/fimmu.2024.1368322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Activation of complement through the alternative pathway (AP) has a key role in the pathogenesis of IgA nephropathy (IgAN). We previously showed, by intraperitoneal injection of Lactobacillus casei cell wall extract (LCWE), C57BL/6 mice develop mild kidney damage in association with glomerular IgA deposition. To further address complement activity in causing glomerular histological alterations as suggested in the pathogenesis of IgAN, here we used mice with factor H mutation (FHW/R) to render AP overactivation in conjunction with LCWE injection to stimulate intestinal production of IgA. Methods Dose response to LCWE were examined between two groups of FHW/R mice. Wild type (FHW/W) mice stimulated with LCWE were used as model control. Results The FHW/R mice primed with high dose LCWE showed elevated IgA and IgA-IgG complex levels in serum. In addition to 100% positive rate of IgA and C3, they display elevated biomarkers of kidney dysfunction, coincided with severe pathological lesions, resembling those of IgAN. As compared to wild type controls stimulated by the same high dose LCWE, these FHW/R mice exhibited stronger complement activation in the kidney and in circulation. Discussion The new mouse model shares many disease features with IgAN. The severity of glomerular lesions and the decline of kidney functions are further aggravated through complement overactivation. The model may be a useful tool for preclinical evaluation of treatment response to complement-inhibitors.
Collapse
Affiliation(s)
- Jingyi Li
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yaping Dong
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Feifei Chen
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hongyu Yang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Pei Chen
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hongyu Li
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Sufang Shi
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xujie Zhou
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Zhu
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuemiao Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Lijun Liu
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xinfang Xie
- Department of Nephrology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Feng Yu
- Department of Nephrology, Peking University International Hospital, Beijing, China
| | - Jing Jin
- Northwestern University Feinberg School of Medicine, Division of Nephrology, Chicago, IL, United States
| | - Jicheng Lv
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
6
|
Sándor N, Schneider AE, Matola AT, Barbai VH, Bencze D, Hammad HH, Papp A, Kövesdi D, Uzonyi B, Józsi M. The human factor H protein family - an update. Front Immunol 2024; 15:1135490. [PMID: 38410512 PMCID: PMC10894998 DOI: 10.3389/fimmu.2024.1135490] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/08/2024] [Indexed: 02/28/2024] Open
Abstract
Complement is an ancient and complex network of the immune system and, as such, it plays vital physiological roles, but it is also involved in numerous pathological processes. The proper regulation of the complement system is important to allow its sufficient and targeted activity without deleterious side-effects. Factor H is a major complement regulator, and together with its splice variant factor H-like protein 1 and the five human factor H-related (FHR) proteins, they have been linked to various diseases. The role of factor H in inhibiting complement activation is well studied, but the function of the FHRs is less characterized. Current evidence supports the main role of the FHRs as enhancers of complement activation and opsonization, i.e., counter-balancing the inhibitory effect of factor H. FHRs emerge as soluble pattern recognition molecules and positive regulators of the complement system. In addition, factor H and some of the FHR proteins were shown to modulate the activity of immune cells, a non-canonical function outside the complement cascade. Recent efforts have intensified to study factor H and the FHRs and develop new tools for the distinction, quantification and functional characterization of members of this protein family. Here, we provide an update and overview on the versatile roles of factor H family proteins, what we know about their biological functions in healthy conditions and in diseases.
Collapse
Affiliation(s)
- Noémi Sándor
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
- HUN-REN-ELTE Complement Research Group, Hungarian Research Network, Budapest, Hungary
| | | | | | - Veronika H. Barbai
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dániel Bencze
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Hani Hashim Hammad
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Alexandra Papp
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dorottya Kövesdi
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
- HUN-REN-ELTE Complement Research Group, Hungarian Research Network, Budapest, Hungary
| | - Barbara Uzonyi
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
- HUN-REN-ELTE Complement Research Group, Hungarian Research Network, Budapest, Hungary
| | - Mihály Józsi
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
- HUN-REN-ELTE Complement Research Group, Hungarian Research Network, Budapest, Hungary
| |
Collapse
|
7
|
Heurich M, McCluskey G. Complement and coagulation crosstalk - Factor H in the spotlight. Immunobiology 2023; 228:152707. [PMID: 37633063 DOI: 10.1016/j.imbio.2023.152707] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/02/2023] [Accepted: 07/10/2023] [Indexed: 08/28/2023]
Abstract
The immune complement and the coagulation systems are blood-based proteolytic cascades that are activated by pathway-specific triggers, based on protein-protein interactions and enzymatic cleavage reactions. Activation of these systems is finely balanced and controlled through specific regulatory mechanisms. The complement and coagulation systems are generally viewed as distinct, but have common evolutionary origins, and several interactions between these homologous systems have been reported. This complement and coagulation crosstalk can affect activation, amplification and regulatory functions in both systems. In this review, we summarize the literature on coagulation factors contributing to complement alternative pathway activation and regulation and highlight molecular interactions of the complement alternative pathway regulator factor H with several coagulation factors. We propose a mechanism where factor H interactions with coagulation factors may contribute to both complement and coagulation activation and regulation within the haemostatic system and fibrin clot microenvironment and introduce the emerging role of factor H as a modulator of coagulation. Finally, we discuss the potential impact of these protein interactions in diseases associated with factor H dysregulation or deficiency as well as evidence of coagulation dysfunction.
Collapse
Affiliation(s)
- Meike Heurich
- School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, United Kingdom.
| | - Geneviève McCluskey
- Université Paris-Saclay, INSERM, Hémostase, Inflammation, Thrombose HITH U1176, 94276 Le Kremlin-Bicêtre, France
| |
Collapse
|
8
|
Mack KL, Talbott HE, Griffin MF, Parker JBL, Guardino NJ, Spielman AF, Davitt MF, Mascharak S, Downer M, Morgan A, Valencia C, Akras D, Berger MJ, Wan DC, Fraser HB, Longaker MT. Allele-specific expression reveals genetic drivers of tissue regeneration in mice. Cell Stem Cell 2023; 30:1368-1381.e6. [PMID: 37714154 PMCID: PMC10592051 DOI: 10.1016/j.stem.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 06/16/2023] [Accepted: 08/22/2023] [Indexed: 09/17/2023]
Abstract
In adult mammals, skin wounds typically heal by scarring rather than through regeneration. In contrast, "super-healer" Murphy Roths Large (MRL) mice have the unusual ability to regenerate ear punch wounds; however, the molecular basis for this regeneration remains elusive. Here, in hybrid crosses between MRL and non-regenerating mice, we used allele-specific gene expression to identify cis-regulatory variation associated with ear regeneration. Analyzing three major cell populations (immune, fibroblast, and endothelial), we found that genes with cis-regulatory differences specifically in fibroblasts were associated with wound-healing pathways and also co-localized with quantitative trait loci for ear wound-healing. Ectopic treatment with one of these proteins, complement factor H (CFH), accelerated wound repair and induced regeneration in typically fibrotic wounds. Through single-cell RNA sequencing (RNA-seq), we observed that CFH treatment dramatically reduced immune cell recruitment to wounds, suggesting a potential mechanism for CFH's effect. Overall, our results provide insights into the molecular drivers of regeneration with potential clinical implications.
Collapse
Affiliation(s)
- Katya L Mack
- Stanford University, Department of Biology, Stanford, CA, USA
| | - Heather E Talbott
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Michelle F Griffin
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Jennifer B L Parker
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Nicholas J Guardino
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Amanda F Spielman
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Michael F Davitt
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Shamik Mascharak
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Mauricio Downer
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Annah Morgan
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Caleb Valencia
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Deena Akras
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Mark J Berger
- Stanford University, Department of Computer Science, Stanford, CA 94305, USA
| | - Derrick C Wan
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Hunter B Fraser
- Stanford University, Department of Biology, Stanford, CA, USA.
| | - Michael T Longaker
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA.
| |
Collapse
|
9
|
Pisani F, Pisani V, Arcangeli F, Harding A, Singhrao SK. Treponema denticola Has the Potential to Cause Neurodegeneration in the Midbrain via the Periodontal Route of Infection-Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6049. [PMID: 37297653 PMCID: PMC10252855 DOI: 10.3390/ijerph20116049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/30/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and the most common example of dementia. The neuropathological features of AD are the abnormal deposition of extracellular amyloid-β (Aβ) and intraneuronal neurofibrillary tangles with hyperphosphorylated tau protein. It is recognized that AD starts in the frontal cerebral cortex, and then it progresses to the entorhinal cortex, the hippocampus, and the rest of the brain. However, some studies on animals suggest that AD could also progress in the reverse order starting from the midbrain and then spreading to the frontal cortex. Spirochetes are neurotrophic: From a peripheral route of infection, they can reach the brain via the midbrain. Their direct and indirect effect via the interaction of their virulence factors and the microglia potentially leads to the host peripheral nerve, the midbrain (especially the locus coeruleus), and cortical damage. On this basis, this review aims to discuss the hypothesis of the ability of Treponema denticola to damage the peripheral axons in the periodontal ligament, to evade the complemental pathway and microglial immune response, to determine the cytoskeletal impairment and therefore causing the axonal transport disruption, an altered mitochondrial migration and the consequent neuronal apoptosis. Further insights about the central neurodegeneration mechanism and Treponema denticola's resistance to the immune response when aggregated in biofilm and its quorum sensing are suggested as a pathogenetic model for the advanced stages of AD.
Collapse
Affiliation(s)
- Flavio Pisani
- Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston PR1 2HE, UK
| | - Valerio Pisani
- IRCCS, “Santa Lucia” Foundation, Neurology and Neurorehabilitation Unit, Via Ardeatina, 306, 00179 Rome, Italy
| | - Francesca Arcangeli
- Azienda Sanitaria Locale ASLRM1, Nuovo Regina Margherita Hospital, Geriatric Department, Advanced Centre for Dementia and Cognitive Disorders, Via Emilio Morosini, 30, 00153 Rome, Italy
| | - Alice Harding
- Dementia and Neurodegenerative Disease Research Group, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston PR1 2HE, UK
| | - Simarjit Kaur Singhrao
- Dementia and Neurodegenerative Disease Research Group, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston PR1 2HE, UK
| |
Collapse
|
10
|
Turner NA, Moake JL. Heat-inactivated Factor B inhibits alternative pathway fluid-phase activation and convertase formation on endothelial cell-secreted ultra-large von Willebrand factor strings. Sci Rep 2023; 13:5764. [PMID: 37031266 PMCID: PMC10082794 DOI: 10.1038/s41598-023-33007-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/05/2023] [Indexed: 04/10/2023] Open
Abstract
Defective regulation of the alternative complement pathway (AP) causes excessive activation and promotes the inflammation and renal injury observed in atypical hemolytic-uremic syndrome (aHUS). The usefulness of heat-inactivated Factor B (HFB) in reducing AP activation was evaluated in: fluid-phase reactions, using purified complement proteins and Factor H (FH)-depleted serum; and in surface-activated reactions using human endothelial cells (ECs). C3a and Ba levels, measured by quantitative Western blots, determined the extent of fluid-phase activation. In reactions using C3, FB, and Factor D proteins, HFB addition (2.5-fold FB levels), reduced C3a levels by 60% and Ba levels by 45%. In reactions using FH-depleted serum (supplemented with FH at 12.5% normal levels), Ba levels were reduced by 40% with HFB added at 3.5-fold FB levels. The effectiveness of HFB in limiting AP convertase formation on activated surfaces was evaluated using stimulated ECs. Fluorescent microscopy was used to quantify endogenously released C3, FB, and C5 attached to EC-secreted ultra-large VWF strings. HFB addition reduced attachment of C3b by 2.7-fold, FB by 1.5-fold and C5 by fourfold. Our data indicate that HFB may be of therapeutic value in preventing AP-mediated generation of C3a and C5a, and the associated inflammation caused by an overactive AP.
Collapse
Affiliation(s)
- Nancy A Turner
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Joel L Moake
- Department of Bioengineering, Rice University, Houston, TX, USA
| |
Collapse
|
11
|
van Essen MF, Peereboom ETM, Schlagwein N, van Gijlswijk-Janssen DJ, Nelemans T, Joeloemsingh JV, van den Berg CW, Prins J, Clark SJ, Schmidt CQ, Trouw LA, van Kooten C. Preferential production and secretion of the complement regulator factor H-like protein 1 (FHL-1) by human myeloid cells. Immunobiology 2023; 228:152364. [PMID: 36881973 DOI: 10.1016/j.imbio.2023.152364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
Factor H is a pivotal complement regulatory protein that is preferentially produced by the liver and circulates in high concentrations in serum. There has been an increasing interest in the extrahepatic production of complement factors, including by cells of the immune system, since this contributes to non-canonical functions of local complement activation and regulation. Here we investigated the production and regulation of factor H and its splice variant factor H-like protein 1 (FHL-1) by human myeloid cells. As validation, we confirmed the predominant presence of intact factor H in serum, despite a strong but comparable mRNA expression of CFH and FHL1 in liver. Comparable levels of CFH and FHL1 were also observed in renal tissue, although a dominant staining for FHL-1 was shown within the proximal tubules. Human in vitro generated pro- and anti-inflammatory macrophages both expressed and produced factor H/FHL-1, but this was strongest in pro-inflammatory macrophages. Production was not affected by LPS activation, but was increased upon stimulation with IFN-γ or CD40L. Importantly, in both macrophage subsets mRNA expression of FHL1 was significantly higher than CFH. Moreover, production of FHL-1 protein could be confirmed using precipitation and immunoblotting of culture supernatants. These data identify macrophages as producers of factor H and FHL-1, thereby potentially contributing to local complement regulation at sites of inflammation.
Collapse
Affiliation(s)
- Mieke F van Essen
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Emma T M Peereboom
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Nicole Schlagwein
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Daniëlle J van Gijlswijk-Janssen
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Tessa Nelemans
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jivan V Joeloemsingh
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Cathelijne W van den Berg
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jurriën Prins
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Simon J Clark
- Institute for Ophthalmic Research, Eberhard Karls University of Tübingen, Tübingen, Baden-Württemberg 72076, Germany; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Christoph Q Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Cees van Kooten
- Division of Nephrology and Transplant Medicine, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
12
|
CFH-CFHR1 hybrid genes in two cases of atypical hemolytic uremic syndrome. J Hum Genet 2023; 68:427-430. [PMID: 36755127 DOI: 10.1038/s10038-023-01129-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023]
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare complement-mediated disease that manifests as the triad of thrombotic microangiopathy. We identified two aHUS patients with neither anti-complement factor H (CFH) antibodies nor causative variants of seven aHUS-related genes (CFH, CFI, CFB, C3, MCP, THBD, and DGKE); however, their plasma showed increased levels of hemolysis by hemolytic assay, which strongly suggests CFH-related abnormalities. Using a copy number variation (CNV) analysis of the CFH/CFHR gene cluster, we identified CFH-CFHR1 hybrid genes in these patients. We verified the absence of aHUS-related abnormal CNVs of the CFH gene in control genomes of 2036 individuals in the general population, which suggests that pathogenicity is related to these hybrid genes. Our study emphasizes that, for patients suspected of having aHUS, it is important to perform an integrated analysis based on a clinical examination, functional analysis, and detailed genetic investigation.
Collapse
|
13
|
Piras R, Valoti E, Alberti M, Bresin E, Mele C, Breno M, Liguori L, Donadelli R, Rigoldi M, Benigni A, Remuzzi G, Noris M. CFH and CFHR structural variants in atypical Hemolytic Uremic Syndrome: Prevalence, genomic characterization and impact on outcome. Front Immunol 2023; 13:1011580. [PMID: 36793547 PMCID: PMC9923232 DOI: 10.3389/fimmu.2022.1011580] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/28/2022] [Indexed: 01/31/2023] Open
Abstract
Introduction Atypical hemolytic uremic syndrome (aHUS) is a rare disease that manifests with microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure, and is associated with dysregulation of the alternative complement pathway. The chromosomal region including CFH and CFHR1-5 is rich in repeated sequences, favoring genomic rearrangements that have been reported in several patients with aHUS. However, there are limited data on the prevalence of uncommon CFH-CFHR genomic rearrangements in aHUS and their impact on disease onset and outcomes. Methods In this study, we report the results of CFH-CFHR Copy Number Variation (CNV) analysis and the characterization of resulting structural variants (SVs) in a large cohort of patients, including 258 patients with primary aHUS and 92 with secondary forms. Results We found uncommon SVs in 8% of patients with primary aHUS: 70% carried rearrangements involving CFH alone or CFH and CFHR (group A; n=14), while 30% exhibited rearrangements including only CFHRs (group B; n=6). In group A, 6 patients presented CFH::CFHR1 hybrid genes, 7 patients carried duplications in the CFH-CFHR region that resulted either in the substitution of the last CFHR1 exon(s) with those of CFH (CFHR1::CFH reverse hybrid gene) or in an internal CFH duplication. In group A, the large majority of aHUS acute episodes not treated with eculizumab (12/13) resulted in chronic ESRD; in contrast, anti-complement therapy induced remission in 4/4 acute episodes. aHUS relapse occurred in 6/7 grafts without eculizumab prophylaxis and in 0/3 grafts with eculizumab prophylaxis. In group B, 5 subjects had the CFHR31-5::CFHR410 hybrid gene and one had 4 copies of CFHR1 and CFHR4. Compared with group A, patients in group B exhibited a higher prevalence of additional complement abnormalities and earlier disease onset. However, 4/6 patients in this group underwent complete remission without eculizumab treatment. In secondary forms we identified uncommon SVs in 2 out of 92 patients: the CFHR31-5::CFHR410 hybrid and a new internal duplication of CFH. Discussion In conclusion, these data highlight that uncommon CFH-CFHR SVs are frequent in primary aHUS and quite rare in secondary forms. Notably, genomic rearrangements involving the CFH are associated with a poor prognosis but carriers respond to anti-complement therapy.
Collapse
|
14
|
Outer surface protein E (OspE) mediates Borrelia burgdorferi sensu stricto strain-specific complement evasion in the eastern fence lizard, Sceloporus undulatus. Ticks Tick Borne Dis 2023; 14:102081. [PMID: 36403322 DOI: 10.1016/j.ttbdis.2022.102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022]
Abstract
In North America, Lyme disease is primarily caused by the spirochetal bacterium Borrelia burgdorferi sensu stricto (Bb), which is transmitted between multiple vertebrate hosts and ixodid ticks, and is a model commonly used to study host-pathogen interactions. While Bb is consistently observed in its mammalian and avian reservoirs, the bacterium is rarely isolated from North American reptiles. Two closely related lizard species, the eastern fence lizard (Sceloporus undulatus) and the western fence lizard (Sceloporus occidentalis), are examples of reptiles parasitized by Ixodes ticks. Vertebrates are known to generate complement as an innate defense mechanism, which can be activated before Bb disseminate to distal tissues. Complement from western fence lizards has proven lethal against one Bb strain, implying the role of complement in making those lizards unable to serve as hosts to Bb. However, Bb DNA is occasionally identified in distal tissues of field-collected eastern fence lizards, suggesting some Bb strains may overcome complement-mediated clearance in these lizards. These findings raise questions regarding the role of complement and its impact on Bb interactions with North American lizards. In this study, we found Bb seropositivity in a small population of wild-caught eastern fence lizards and observed Bb strain-specific survivability in lizard sera. We also found that a Bb outer surface protein, OspE, from Bb strains viable in sera, promotes lizard serum survivability and binds to a complement inhibitor, factor H, from eastern fence lizards. Our data thus identify bacterial and host determinants of eastern fence lizard complement evasion, providing insights into the role of complement influencing Bb interactions with North American lizards.
Collapse
|
15
|
Rodríguez de Córdoba S. Genetic variability shapes the alternative pathway complement activity and predisposition to complement-related diseases. Immunol Rev 2023; 313:71-90. [PMID: 36089777 PMCID: PMC10086816 DOI: 10.1111/imr.13131] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The implementation of next-generation sequencing technologies has provided a sharp picture of the genetic variability in the components and regulators of the alternative pathway (AP) of the complement system and has revealed the association of many AP variants with different rare and common diseases. An important finding that has emerged from these analyses is that each of these complement-related diseases associate with genetic variants altering specific aspects of the activation and regulation of the AP. These genotype-phenotype correlations have provided valuable insights into their pathogenic mechanisms with important diagnostic and therapeutic implications. While genetic variants in coding regions and structural variants are reasonably well characterized and occasionally have been instrumental to uncover unknown features of the complement proteins, data about complement expressed quantitative trait loci are still very limited. A crucial task for future studies will be to identify these quantitative variations and to determine their impact in the overall activity of the AP. This is fundamental as it is now clear that the consequences of genetic variants in the AP are additive and that susceptibility or resistance to disease is the result of specific combinations of genetic variants in different complement components and regulators ("complotypes").
Collapse
|
16
|
Xiao Z, Yeung CLS, Yam JWP, Mao X. An update on the role of complement in hepatocellular carcinoma. Front Immunol 2022; 13:1007382. [PMID: 36341431 PMCID: PMC9629811 DOI: 10.3389/fimmu.2022.1007382] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
As a main producer of complement, the environment in the liver is greatly affected by the complement system. Although the complement system is considered to have the ability of nonself discrimination, remarkable studies have revealed the tight association between improper complement activation in tumour initiation and progression. As complement activation predominantly occurs within the liver, the protumourigenic role of the complement system may contribute to the development of hepatocellular carcinoma (HCC). Improvement in the understanding of the molecular targets involved in complement-mediated tumour development, metastasis, and tumour-promoting inflammation in HCC would certainly aid in the development of better treatments. This minireview is focused on recent findings of the protumourigenic role of the complement system in HCC.
Collapse
Affiliation(s)
- Zhijie Xiao
- Scientific Research Centre, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Charlie Lot Sum Yeung
- Department of Pathology, School of Clinical Medicine, Faculty of Medicine, the University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Faculty of Medicine, the University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiaowen Mao
- Department of Pathology, School of Clinical Medicine, Faculty of Medicine, the University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Xiaowen Mao,
| |
Collapse
|
17
|
Abstract
Neisseria meningitidis and Neisseria gonorrhoeae are important human pathogens that have evolved to bind the major negative regulator of the complement system, complement factor H (CFH). However, little is known about the interaction of pathogens with CFH-related proteins (CFHRs) which are structurally similar to CFH but lack the main complement regulatory domains found in CFH. Insights into the role of CFHRs have been hampered by a lack of specific reagents. We generated a panel of CFHR-specific monoclonal antibodies and demonstrated that CFHR5 was bound by both pathogenic Neisseria spp. We showed that CFHR5 bound to PorB expressed by both pathogens in the presence of sialylated lipopolysaccharide and enhanced complement activation on the surface of N. gonorrhoeae. Our study furthered our understanding of the interactions of CFHRs with bacterial pathogens and revealed that CFHR5 bound the meningococcus and gonococcus via similar mechanisms.
Collapse
|
18
|
Kumar V, Pouw RB, Autio MI, Sagmeister MG, Phua ZY, Borghini L, Wright VJ, Hoggart C, Pan B, Tan AKY, Binder A, Brouwer MC, Pinnock E, De Groot R, Hazelzet J, Emonts M, Van Der Flier M, Reiter K, Nöthen MM, Hoffmann P, Schlapbach LJ, Bellos E, Anderson S, Secka F, Martinón-Torres F, Salas A, Fink C, Carrol ED, Pollard AJ, Coin LJ, Zenz W, Wouters D, Ang LT, Hibberd ML, Levin M, Kuijpers TW, Davila S. Variation in CFHR3 determines susceptibility to meningococcal disease by controlling factor H concentrations. Am J Hum Genet 2022; 109:1680-1691. [PMID: 36007525 PMCID: PMC9502058 DOI: 10.1016/j.ajhg.2022.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/31/2022] [Indexed: 11/16/2022] Open
Abstract
Neisseria meningitidis protects itself from complement-mediated killing by binding complement factor H (FH). Previous studies associated susceptibility to meningococcal disease (MD) with variation in CFH, but the causal variants and underlying mechanism remained unknown. Here we attempted to define the association more accurately by sequencing the CFH-CFHR locus and imputing missing genotypes in previously obtained GWAS datasets of MD-affected individuals of European ancestry and matched controls. We identified a CFHR3 SNP that provides protection from MD (rs75703017, p value = 1.1 × 10-16) by decreasing the concentration of FH in the blood (p value = 1.4 × 10-11). We subsequently used dual-luciferase studies and CRISPR gene editing to establish that deletion of rs75703017 increased FH expression in hepatocyte by preventing promotor inhibition. Our data suggest that reduced concentrations of FH in the blood confer protection from MD; with reduced access to FH, N. meningitidis is less able to shield itself from complement-mediated killing.
Collapse
Affiliation(s)
- Vikrant Kumar
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore; Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Richard B Pouw
- Division of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, the Netherlands; Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - Matias I Autio
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore; Cardiovascular Research Institute, Centre for Translational Medicine, National University Health System, Singapore
| | | | - Zai Yang Phua
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - Lisa Borghini
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore; Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain; GenPoB Research Group, Instituto de Investigación Sanitaria de Santiago, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Victoria J Wright
- Section of Paediatric Infectious Disease, Division of Infectious Disease, Department of Medicine, Imperial College London, London, UK
| | - Clive Hoggart
- Section of Paediatric Infectious Disease, Division of Infectious Disease, Department of Medicine, Imperial College London, London, UK
| | - Bangfen Pan
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore; Cardiovascular Research Institute, Centre for Translational Medicine, National University Health System, Singapore
| | - Antson Kiat Yee Tan
- Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Alexander Binder
- Department of General Paediatrics, Medical University of Graz, Graz, Austria
| | - Mieke C Brouwer
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | | - Ronald De Groot
- Section of Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan Hazelzet
- Department of Pediatrics, Erasmus Medical Center-Sophia Children's Hospital, University Medical Center, Rotterdam, the Netherlands
| | - Marieke Emonts
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK; National Institute for Health and Care Research Newcastle Biomedical Research Centre Based at Newcastle Upon Tyne Hospitals National Health Service Trust and Newcastle University, Newcastle Upon Tyne, UK; Paediatric Infectious Diseases and Immunology Department, Newcastle Upon Tyne Hospitals Foundation Trust, Great North Children's Hospital, Newcastle Upon Tyne, UK
| | - Michiel Van Der Flier
- Section of Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Paediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Karl Reiter
- Department of Paediatrics, Division of Paediatric Intensive Care Medicine, Ludwig Maximilian University of Munich and Dr. von Hauner's Children's Hospital, Munich, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | | | - Luregn J Schlapbach
- Child Health Research Centre, The University of Queensland, Brisbane, Australia; Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, Australia; Department of Intensive Care and Neonatology and Children`s Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Evangelos Bellos
- Section of Paediatric Infectious Disease, Division of Infectious Disease, Department of Medicine, Imperial College London, London, UK
| | | | - Fatou Secka
- Medical Research Council Unit Gambia, Banjul, The Gambia
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain; Genetics, Vaccines, Infectious Diseases, and Pediatrics Research Group, Instituto de Investigación Sanitaria de Santiago, Universidad de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Salas
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain; GenPoB Research Group, Instituto de Investigación Sanitaria de Santiago, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Colin Fink
- Micropathology, University of Warwick, Coventry, UK
| | - Enitan D Carrol
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Lachlan J Coin
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Werner Zenz
- Department of General Paediatrics, Medical University of Graz, Graz, Austria
| | - Diana Wouters
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - Lay Teng Ang
- Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Martin L Hibberd
- Infectious Diseases, Genome Institute of Singapore, Singapore, Singapore; Infectious and Tropical Disease, London School of Hygiene & Tropical Medicine, London, UK
| | - Michael Levin
- Section of Paediatric Infectious Disease, Division of Infectious Disease, Department of Medicine, Imperial College London, London, UK
| | - Taco W Kuijpers
- Division of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, the Netherlands.
| | - Sonia Davila
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore; Duke-National University of Singapore Medical School, Singapore, Singapore; SingHealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore.
| |
Collapse
|
19
|
Lee YJ, Lin YC, Liao CC, Chang YS, Huang YH, Tsai IJ, Chen JH, Lin SH, Lin YF, Hsieh TW, Chen YS, Wu CY, Chang CC, Lin CY. Using anti-malondialdehyde-modified peptide adduct autoantibodies in serum of taiwanese women to diagnose primary Sjogren's syndrome. Clin Biochem 2022; 108:27-41. [PMID: 35843269 DOI: 10.1016/j.clinbiochem.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/20/2022] [Accepted: 07/11/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Sjogren's syndrome (SS) is a systemic autoimmune disease featured with a dry mouth and dry eyes. Several autoantibodies, including anti-SSA, anti-SSB, antinuclear antibodies can be detected in patients with SS. Oxidation-specific epitopes (OSEs) can be formed from malondialdehyde (MDA)-modified protein adducts and trigger chronic inflammation. In this study, our purposes were used serum levels of anti-MDA-modified peptide adducts autoantibodies to evaluate predictive performance by machine learning algorithms in primary Sjögren's syndrome (pSS) and assess the association between pSS and healthy controls. METHODS Three novel MDA-modified peptide adducts, including immunoglobulin (Ig) gamma heavy chain 1 (IGHG1)102-131, complement factor H (CFAH)1045-1062, and Ig heavy constant alpha 1 (IGHA1)307-327 were identified and validated. Serum levels of protein, MDA-modified protein adducts, MDA, and autoantibodies recognizing unmodified peptides and MDA-modified peptide adducts were measured. Statistically significance in correlations and odds ratios (ORs) were estimated. RESULTS The random forest classifier utilized autoantibodies combination composed of IgM anti-IGHG1102-131, IgM anti-IGHG1102-131 MDA and IgM anti-IGHA1307-327 achieved predictive performance as an accuracy of 88.0%, a sensitivity of 93.7%, and a specificity of 84.4% which may be as potential diagnostic biomarkers to differentiate patients with pSS from rheumatoid arthritis (RA), and secondary SS in RA and HCs. CONCLUSIONS Our findings imply that low levels of IgA anti-IGHG1102-131 MDA (OR = 2.646), IgA anti-IGHG1102-131 (OR = 2.408), IgA anti-CFAH1045-1062 (OR = 2.571), and IgA anti-IGHA1307-327 (OR = 2.905) may denote developing risks of pSS, respectively.
Collapse
Affiliation(s)
- Yuarn-Jang Lee
- Division of Infectious Diseases, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan; Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ying-Chin Lin
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Geriatric Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chen-Chung Liao
- Proteomics Research Center, National Yang-Ming University, Taipei 112, Taiwan
| | - Yu-Sheng Chang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Hui Huang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - I-Jung Tsai
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Jin-Hua Chen
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei 11031, Taiwan; Statistics Center, Office of Data Science, Taipei Medical University, Taipei 11031, Taiwan
| | - Sheng-Hong Lin
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Fang Lin
- Department of Laboratory Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Ting-Wan Hsieh
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Department of Laboratory Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Yi-Su Chen
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chih-Yin Wu
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chi-Ching Chang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Ching-Yu Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
20
|
Zhu Q, Wang J, Zhang W, Zhu W, Wu Z, Chen Y, Chen M, Zheng L, Tang J, Zhang S, Wang D, Wang X, Chen G. Whole-Genome/Exome Sequencing Uncovers Mutations and Copy Number Variations in Primary Diffuse Large B-Cell Lymphoma of the Central Nervous System. Front Genet 2022; 13:878618. [PMID: 35646048 PMCID: PMC9133733 DOI: 10.3389/fgene.2022.878618] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
Background/objective: Identification of key genetic alterations is of importance in the targeted therapies of primary central nervous system lymphoma (PCNSL). However, only a small number of studies have been carried out in PCNSL. In this study, we further described the genetic mutations and copy number variations (CNVs) in PCNSL patients using whole-genome/exome sequencing (WGS/WES), as well as revealed their associations with patients’ clinicopathological features and prognosis. Methods: Tumor specimens from 38 patients with primary diffuse large B-cell lymphoma of the central nervous system (CNS DLBCL) were enrolled to WGS (n = 24) or WES (n = 14). The CNVs and mutations of 24 samples (WGS) and 38 samples (WGS/WES) were characterized, respectively. The associations between CNVs and mutations with the overall survival rates of PCNSL patients were also evaluated. Results: The most common mutations were identified in IGLL5 (68%), PIM1 (63%), MYD88 (55%), CD79B (42%), BTG2 (39%), PCLO (39%), KMT2D (34%), and BTG1 (29%) genes. Among the mutated genes, EP300, ETV6, and HIST1H1E mutations were exclusively detected in the elderly, while DUSP2 mutations were associated with the immune microenvironment indicators. In addition, KMT2D mutation was associated with a poor prognosis. In addition, 488 CNVs including 91 gains and 397 deletions were observed across 24 samples from WGS results. Notably, 1q31.3 amplification was closely associated with the poor prognosis of PCNSL patients. Conclusion: This study further characterizes the genomic landscape of primary CNS DLBCL using WGS/WES, which provides insight into understanding the pathogenesis of PCNSL and fosters new ideas for the targeted treatment of PCNSL.
Collapse
Affiliation(s)
- Qiong Zhu
- Department of Molecular Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jianchao Wang
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Wenfang Zhang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Weifeng Zhu
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Zaizeng Wu
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yanping Chen
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Musheng Chen
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Limei Zheng
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jianqing Tang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Di Wang
- Department of Molecular Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Xingfu Wang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Gang Chen
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| |
Collapse
|
21
|
Kiss MG, Binder CJ. The multifaceted impact of complement on atherosclerosis. Atherosclerosis 2022; 351:29-40. [DOI: 10.1016/j.atherosclerosis.2022.03.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/17/2022] [Accepted: 03/09/2022] [Indexed: 12/12/2022]
|
22
|
Essen MF, Schlagwein N, den Hoven EM, Gijlswijk‐Janssen DJ, Lubbers R, den Bos RM, den Born J, Ruben JM, Trouw LA, Kooten C. Initial properdin binding contributes to alternative pathway activation at the surface of viable and necrotic cells. Eur J Immunol 2022; 52:597-608. [PMID: 35092629 PMCID: PMC9303752 DOI: 10.1002/eji.202149259] [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] [Received: 03/16/2021] [Revised: 12/01/2021] [Accepted: 01/10/2022] [Indexed: 11/16/2022]
Abstract
Properdin, the only known positive regulator of the complement system, stabilizes the C3 convertase, thereby increasing its half‐life. In contrast to most other complement factors, properdin is mainly produced extrahepatically by myeloid cells. Recent data suggest a role for properdin as a pattern recognition molecule. Here, we confirmed previous findings of properdin binding to different necrotic cells including Jurkat T cells. Binding can occur independent of C3, as demonstrated by HAP‐1 C3 KO cells, excluding a role for endogenous C3. In view of the cellular source of properdin, interaction with myeloid cells was examined. Properdin bound to the surface of viable monocyte‐derived pro‐ and anti‐inflammatory macrophages, but not to DCs. Binding was demonstrated for purified properdin as well as fractionated P2, P3, and P4 properdin oligomers. Binding contributed to local complement activation as determined by C3 and C5b‐9 deposition on the cell surfaces and seems a prerequisite for alternative pathway activation. Interaction of properdin with cell surfaces could be inhibited with the tick protein Salp20 and by different polysaccharides, depending on sulfation and chain length. These data identify properdin as a factor interacting with different cell surfaces, being either dead or alive, contributing to the local stimulation of complement activation.
Collapse
Affiliation(s)
- Mieke F. Essen
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Nicole Schlagwein
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Elisa M.P. den Hoven
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Daniëlle J. Gijlswijk‐Janssen
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Rosalie Lubbers
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
| | - Ramon M. den Bos
- Crystal and Structural Chemistry Bijvoet Center for Biomolecular Research Department of Chemistry Faculty of Science Utrecht University Utrecht The Netherlands
| | - Jacob den Born
- Department of Nephrology University Medical Center Groningen Groningen The Netherlands
| | - Jurjen M. Ruben
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Leendert A. Trouw
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
- Department of Immunology Leiden University Medical Center Leiden The Netherlands
| | - Cees Kooten
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | | |
Collapse
|
23
|
Xu B, Kang Y, Du Y, Guo W, Zhu L, Zhang H. Atypical Hemolytic Uremic Syndrome-Associated FHR1 Isoform FHR1*B Enhances Complement Activation and Inflammation. Front Immunol 2022; 13:755694. [PMID: 35126388 PMCID: PMC8814109 DOI: 10.3389/fimmu.2022.755694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare but severe type of thrombotic microangiopathy that is triggered by the abnormal activation of the alternative complement pathway. Previous studies have reported that three completely linked coding variants of CFHR1 form two haplotypes, namely, CFHR1*A (c.469C, c.475C, c.523G) and CFHR1*B (c.469T, c.475G, c.523C). CFHR1*B is associated with susceptibility to aHUS. To explore the genetic mechanism by which CFHR1 isoforms contribute to aHUS, we compared the structures of FHR1*A and FHR1*B by homology modeling and found differences in the angles between SCR3 and SCR4-SCR5, as FHR1*B had a larger angle than FHR1*A. Then, we expressed FHR1*A and FHR1*B recombinant proteins and compared their functions in complement system regulation and inflammation. We found that FHR1*B presented a significantly higher capacity for binding C3b and necrotic cells than FHR1*A. In a cofactor assay, the FHR-1*B showed stronger influence on FH mediated cofactor function than the FHR-1*A, resulted in fewer C3b cleavage products. In the C3 convertase assays, FHR1*B showed more powerful effect compared with FHR1*A regarding to de-regulate FH function of inhibition the assembling of C3bBb. Additionally, we also found that FHR1*B triggered monocytes to secrete higher levels of IL-1β and IL-6 than FHR1*A. In the present study, we showed that variants of CFHR1 might differently affect complement activation and sterile inflammation. Our findings provide a possible mechanism underlying the predisposition to aHUS caused by CFHR1 isoform CFHR1*B.
Collapse
Affiliation(s)
- Boyang Xu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease (Peking University), National Health Commission, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
| | - Yuqi Kang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease (Peking University), National Health Commission, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
| | - Yujing Du
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Weiyi Guo
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease (Peking University), National Health Commission, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
| | - Li Zhu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease (Peking University), National Health Commission, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease (Peking University), National Health Commission, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
| |
Collapse
|
24
|
Kamala O, Malik TH, Hallam TM, Cox TE, Yang Y, Vyas F, Luli S, Connelly C, Gibson B, Smith-Jackson K, Denton H, Pappworth IY, Huang L, Kavanagh D, Pickering MC, Marchbank KJ. Homodimeric Minimal Factor H: In Vivo Tracking and Extended Dosing Studies in Factor H Deficient Mice. Front Immunol 2021; 12:752916. [PMID: 34956184 PMCID: PMC8696033 DOI: 10.3389/fimmu.2021.752916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway (AP) of complement and treatment options remain inadequate. Factor H (FH) is a potent regulator of the AP. An in-depth analysis of FH-related protein dimerised minimal (mini)-FH constructs has recently been published. This analysis showed that addition of a dimerisation module to mini-FH not only increased serum half-life but also improved complement regulatory function, thus providing a potential treatment option for C3G. Herein, we describe the production of a murine version of homodimeric mini-FH [mHDM-FH (mFH1-5^18-20^R1-2)], developed to reduce the risk of anti-drug antibody formation during long-term experiments in murine models of C3G and other complement-driven pathologies. Our analysis of mHDM-FH indicates that it binds with higher affinity and avidity to WT mC3b when compared to mouse (m)FH (mHDM-FH KD=505 nM; mFH KD=1370 nM) analogous to what we observed with the respective human proteins. The improved binding avidity resulted in enhanced complement regulatory function in haemolytic assays. Extended interval dosing studies in CFH-/- mice (5mg/kg every 72hrs) were partially effective and bio-distribution analysis in CFH-/- mice, through in vivo imaging technologies, demonstrates that mHDM-FH is preferentially deposited and remains fixed in the kidneys (and liver) for up to 4 days. Extended dosing using an AAV- human HDM-FH (hHDM-FH) construct achieved complete normalisation of C3 levels in CFH-/- mice for 3 months and was associated with a significant reduction in glomerular C3 staining. Our data demonstrate the ability of gene therapy delivery of mini-FH constructs to enhance complement regulation in vivo and support the application of this approach as a novel treatment strategy in diseases such as C3G.
Collapse
Affiliation(s)
- Ola Kamala
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Talat H. Malik
- Centre for Inflammatory Disease, Imperial College London, London, United Kingdom
| | - Thomas M. Hallam
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Thomas E. Cox
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Yi Yang
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Falguni Vyas
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Saimir Luli
- Preclinical In Vivo Imaging, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Chloe Connelly
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Beth Gibson
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Kate Smith-Jackson
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Harriet Denton
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Isabel Y. Pappworth
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Lei Huang
- Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - David Kavanagh
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Matthew C. Pickering
- Centre for Inflammatory Disease, Imperial College London, London, United Kingdom
| | - Kevin J. Marchbank
- Complement Therapeutics Research Group and National Renal Complement Therapeutics Centre, Translational and Clinical Research Institute, The Medical School, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
25
|
Kárpáti É, Kremlitzka M, Sándor N, Hajnal D, Schneider AE, Józsi M. Complement Factor H Family Proteins Modulate Monocyte and Neutrophil Granulocyte Functions. Front Immunol 2021; 12:660852. [PMID: 34671340 PMCID: PMC8521052 DOI: 10.3389/fimmu.2021.660852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 09/07/2021] [Indexed: 01/13/2023] Open
Abstract
Besides being a key effector arm of innate immunity, a plethora of non-canonical functions of complement has recently been emerging. Factor H (FH), the main regulator of the alternative pathway of complement activation, has been reported to bind to various immune cells and regulate their functions, beyond its role in modulating complement activation. In this study we investigated the effect of FH, its alternative splice product FH-like protein 1 (FHL-1), the FH-related (FHR) proteins FHR-1 and FHR-5, and the recently developed artificial complement inhibitor mini-FH, on two key innate immune cells, monocytes and neutrophilic granulocytes. We found that, similar to FH, the other factor H family proteins FHL-1, FHR-1 and FHR-5, as well as the recombinant mini-FH, are able to bind to both monocytes and neutrophils. As a functional outcome, immobilized FH and FHR-1 inhibited PMA-induced NET formation, but increased the adherence and IL-8 production of neutrophils. FHL-1 increased only the adherence of the cells, while FHR-5 was ineffective in altering these functions. The adherence of monocytes was increased on FH, recombinant mini-FH and FHL-1 covered surfaces and, except for FHL-1, the same molecules also enhanced secretion of the inflammatory cytokines IL-1β and TNFα. When monocytes were stimulated with LPS in the presence of immobilized FH family proteins, FH, FHL-1 and mini-FH enhanced whereas FHR-1 and FHR-5 decreased the secretion of TNFα; FHL-1 and mini-FH also enhanced IL-10 release compared to the effect of LPS alone. Our results reveal heterogeneous effects of FH and FH family members on monocytes and neutrophils, altering key features involved in pathogen killing, and also demonstrate that FH-based complement inhibitors, such as mini-FH, may have effects beyond their function of inhibiting complement activation. Thus, our data provide new insight into the non-canonical functions of FH, FHL-1, FHR-1 and FHR-5 that might be exploited during protection against infections and in vaccine development.
Collapse
Affiliation(s)
- Éva Kárpáti
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Mariann Kremlitzka
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Noémi Sándor
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary.,MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Dávid Hajnal
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Andrea E Schneider
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Mihály Józsi
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary.,MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
26
|
Hart TM, Dupuis AP, Tufts DM, Blom AM, Starkey SR, Rego ROM, Ram S, Kraiczy P, Kramer LD, Diuk-Wasser MA, Kolokotronis SO, Lin YP. Host tropism determination by convergent evolution of immunological evasion in the Lyme disease system. PLoS Pathog 2021; 17:e1009801. [PMID: 34324600 PMCID: PMC8354441 DOI: 10.1371/journal.ppat.1009801] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/10/2021] [Accepted: 07/14/2021] [Indexed: 11/18/2022] Open
Abstract
Pathogens possess the ability to adapt and survive in some host species but not in others-an ecological trait known as host tropism. Transmitted through ticks and carried mainly by mammals and birds, the Lyme disease (LD) bacterium is a well-suited model to study such tropism. Three main causative agents of LD, Borrelia burgdorferi, B. afzelii, and B. garinii, vary in host ranges through mechanisms eluding characterization. By feeding ticks infected with different Borrelia species, utilizing feeding chambers and live mice and quail, we found species-level differences in bacterial transmission. These differences localize on the tick blood meal, and specifically complement, a defense in vertebrate blood, and a polymorphic bacterial protein, CspA, which inactivates complement by binding to a host complement inhibitor, Factor H (FH). CspA selectively confers bacterial transmission to vertebrates that produce FH capable of allele-specific recognition. CspA is the only member of the Pfam54 gene family to exhibit host-specific FH-binding. Phylogenetic analyses revealed convergent evolution as the driver of such uniqueness, and that FH-binding likely emerged during the last glacial maximum. Our results identify a determinant of host tropism in Lyme disease infection, thus defining an evolutionary mechanism that shapes host-pathogen associations.
Collapse
Affiliation(s)
- Thomas M. Hart
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biological Sciences, State University of New York at Albany, Albany, New York, United States of America
| | - Alan P. Dupuis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Danielle M. Tufts
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, New York, United States of America
| | - Anna M. Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmo, Sweden
| | - Simon R. Starkey
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Ryan O. M. Rego
- Institute of Parasitology, Czech Academy of Sciences, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Laura D. Kramer
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, State University of New York at Albany, Albany, New York, United States of America
| | - Maria A. Diuk-Wasser
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, New York, United States of America
| | - Sergios-Orestis Kolokotronis
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, New York, United States of America
- Institute for Genomic Health, SUNY Downstate Health Sciences University, Brooklyn, New York, United States of America
- Division of Infectious Diseases, Department of Medicine, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York, United States of America
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, State University of New York at Albany, Albany, New York, United States of America
| |
Collapse
|
27
|
Losing control to bad relatives. Blood 2021; 137:3462-3463. [PMID: 34165547 DOI: 10.1182/blood.2021011364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
28
|
Moore SR, Menon SS, Cortes C, Ferreira VP. Hijacking Factor H for Complement Immune Evasion. Front Immunol 2021; 12:602277. [PMID: 33717083 PMCID: PMC7947212 DOI: 10.3389/fimmu.2021.602277] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
The complement system is an essential player in innate and adaptive immunity. It consists of three pathways (alternative, classical, and lectin) that initiate either spontaneously (alternative) or in response to danger (all pathways). Complement leads to numerous outcomes detrimental to invaders, including direct killing by formation of the pore-forming membrane attack complex, recruitment of immune cells to sites of invasion, facilitation of phagocytosis, and enhancement of cellular immune responses. Pathogens must overcome the complement system to survive in the host. A common strategy used by pathogens to evade complement is hijacking host complement regulators. Complement regulators prevent attack of host cells and include a collection of membrane-bound and fluid phase proteins. Factor H (FH), a fluid phase complement regulatory protein, controls the alternative pathway (AP) both in the fluid phase of the human body and on cell surfaces. In order to prevent complement activation and amplification on host cells and tissues, FH recognizes host cell-specific polyanionic markers in combination with complement C3 fragments. FH suppresses AP complement-mediated attack by accelerating decay of convertases and by helping to inactivate C3 fragments on host cells. Pathogens, most of which do not have polyanionic markers, are not recognized by FH. Numerous pathogens, including certain bacteria, viruses, protozoa, helminths, and fungi, can recruit FH to protect themselves against host-mediated complement attack, using either specific receptors and/or molecular mimicry to appear more like a host cell. This review will explore pathogen complement evasion mechanisms involving FH recruitment with an emphasis on: (a) characterizing the structural properties and expression patterns of pathogen FH binding proteins, as well as other strategies used by pathogens to capture FH; (b) classifying domains of FH important in pathogen interaction; and (c) discussing existing and potential treatment strategies that target FH interactions with pathogens. Overall, many pathogens use FH to avoid complement attack and appreciating the commonalities across these diverse microorganisms deepens the understanding of complement in microbiology.
Collapse
Affiliation(s)
- Sara R Moore
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Smrithi S Menon
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Claudio Cortes
- Department of Foundational Medical Sciences, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
29
|
Zarantonello A, Pedersen H, Laursen NS, Andersen GR. Nanobodies Provide Insight into the Molecular Mechanisms of the Complement Cascade and Offer New Therapeutic Strategies. Biomolecules 2021; 11:biom11020298. [PMID: 33671302 PMCID: PMC7922070 DOI: 10.3390/biom11020298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 01/22/2023] Open
Abstract
The complement system is part of the innate immune response, where it provides immediate protection from infectious agents and plays a fundamental role in homeostasis. Complement dysregulation occurs in several diseases, where the tightly regulated proteolytic cascade turns offensive. Prominent examples are atypical hemolytic uremic syndrome, paroxysmal nocturnal hemoglobinuria and Alzheimer’s disease. Therapeutic intervention targeting complement activation may allow treatment of such debilitating diseases. In this review, we describe a panel of complement targeting nanobodies that allow modulation at different steps of the proteolytic cascade, from the activation of the C1 complex in the classical pathway to formation of the C5 convertase in the terminal pathway. Thorough structural and functional characterization has provided a deep mechanistic understanding of the mode of inhibition for each of the nanobodies. These complement specific nanobodies are novel powerful probes for basic research and offer new opportunities for in vivo complement modulation.
Collapse
Affiliation(s)
- Alessandra Zarantonello
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (A.Z.); (H.P.)
| | - Henrik Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (A.Z.); (H.P.)
| | - Nick S. Laursen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
| | - Gregers R. Andersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (A.Z.); (H.P.)
- Correspondence: ; Tel.: +45-30256646
| |
Collapse
|
30
|
Tao J, Tan M, Li LL, Chu H, Song D, Tan Y, Wang SX, Qu Z, Yu F. Genetic Variant CFH rs6677604 Might Play a Protective Role in lupus Nephritis. Am J Med Sci 2020; 361:336-343. [PMID: 33309135 DOI: 10.1016/j.amjms.2020.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/08/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND This study aimed to explore the associations between the complement factor H (CFH) rs6677604 and clinico-pathological characteristics of lupus nephritis. MATERIALS AND METHODS A total of 188 patients with lupus nephritis with complete clinico-pathological data were enrolled and genotyping of CFH rs6677604 was conducted by TaqMan SNP genotyping assays. Patients were divided into two groups by rs6677604-AA/AG or -GG, and the clinico-pathological features between the two groups were further compared. RESULTS We found that patients with rs6677604-AA/AG presented with lower prevalence of anti-dsDNA antibody (12/24 [50.0%] vs 121/164 [73.8%], P = 0.028), higher level of plasma C3a (2642.96 ± 1575.05 vs 1640.01 ± 1209.40, ng/ml, P = 0.024), and a tendency for higher level of plasma CFH (505.76 ± 169.28 vs 397.67 ± 179.11, μg/ml, P = 0.087). Patients with rs6677604-AA/AG had milder renal histopathological features, including total activity indices score (4.5[0, 13] vs 8[0, 19], P = 0.013), endocapillary hypercellularity (1.5[0, 3] vs 3[0, 3], P = 0.013), sub-endothelial hyaline deposits (0.5[0, 3] vs 1[0,3], P = 0.021), glomerular leukocyte infiltration (0.5[0, 1] vs 1[0, 12], P = 0.023) and tubular atrophy (1[0, 1] vs 1[0, 3], P = 0.027) than those with rs6677604-GG, which was further confirmed by the stratified analysis. The rs6677604-A was not a risk factor for patients' renal outcomes (hazard ratio=0.898; 95% CI: 0.264-3.059, P = 0.863). CONCLUSIONS The rs6677604-A genotype in CFH was associated with milder renal pathological features in lupus nephritis, and its protective effect on the pathogenesis of the disease remained to be elucidated.
Collapse
Affiliation(s)
- Juan Tao
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Meng Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Lin-Lin Li
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Hong Chu
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Di Song
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - Su-Xia Wang
- Laboratory of Electron Microscopy, Pathological Centre, Peking University First Hospital, Beijing, PR China
| | - Zhen Qu
- Department of Nephrology, Peking University International Hospital, Beijing, PR China.
| | - Feng Yu
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China; Department of Nephrology, Peking University International Hospital, Beijing, PR China
| |
Collapse
|
31
|
Kuroki Y, Mitsuiki K, Nakagawa K, Tsuruya K, Katafuchi R, Hirakata H, Nakano T. Late renal recovery after treatment over 1 year post-onset in an atypical hemolytic uremic syndrome: a case report. BMC Nephrol 2020; 21:236. [PMID: 32571244 PMCID: PMC7310110 DOI: 10.1186/s12882-020-01897-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/17/2020] [Indexed: 11/27/2022] Open
Abstract
Background Atypical hemolytic uremic syndrome (aHUS) is a life-threatening disease that leads to end-stage kidney disease if only a poor response to plasma exchanges (PEs) or eculizumab therapy is achieved. Case presentation A 58-year-old Japanese man presented with thrombocytopenia, anemia, and kidney failure requiring dialysis without any underlying disease. A kidney biopsy revealed marked mesangiolysis in all glomeruli, compatible with thrombotic microangiopathy (TMA). Based on the positive anti- factor H antibody and negative result for secondary TMA, we diagnosed him as aHUS. Despite eculizumab administration after eight sessions of PE, neither platelet normalization nor kidney recovery was achieved. Eight months later, we discontinued eculizumab therapy due to anaphylactic reaction. At 15 months after the onset of TMA, his platelet count increased gradually from 40 to 150 × 103/μL with a decreased serum creatinine level and increased urine output, eventually allowing the withdrawal of dialysis therapy. A second kidney biopsy showed mesangial widening compatible with the healing of TMA. Conclusions This case indicates that aHUS with PEs and eculizumab therapy has the potential for renal recovery even if over 1 year has passed.
Collapse
Affiliation(s)
- Yusuke Kuroki
- Nephrology & Dialysis Center, Japanese Red Cross Fukuoka Hospital, 3-1-1, Ogusu, Minami-ku, Fukuoka, 815-8555, Japan.
| | - Koji Mitsuiki
- Nephrology & Dialysis Center, Japanese Red Cross Fukuoka Hospital, 3-1-1, Ogusu, Minami-ku, Fukuoka, 815-8555, Japan
| | - Kaneyasu Nakagawa
- Nephrology & Dialysis Center, Japanese Red Cross Fukuoka Hospital, 3-1-1, Ogusu, Minami-ku, Fukuoka, 815-8555, Japan
| | | | - Ritsuko Katafuchi
- Division of Nephrology, National Hospital Organization Fukuokahigashi Medical Center, Koga, Japan
| | - Hideki Hirakata
- Nephrology & Dialysis Center, Japanese Red Cross Fukuoka Hospital, 3-1-1, Ogusu, Minami-ku, Fukuoka, 815-8555, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
32
|
Anti-complement factor H autoantibodies may be protective in lupus nephritis. Clin Chim Acta 2020; 508:1-8. [PMID: 32387092 DOI: 10.1016/j.cca.2020.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND This study aimed to investigate the role of anti-CFH autoantibodies in lupus nephritis based on a well-defined cohort. METHODS One hundred twenty patients with biopsy-proven active lupus nephritis were collected as the discovery cohort, sixty patients served as the validation cohort, thirty-four patients with SLE without renal involvement (NR-SLE) were as disease controls, and thirty healthy donors were also included. The anti-CFH autoantibodies and IgG subclasses were detected by ELISA, and epitopes were evaluated by western blot. Anti-CFH autoantibodies were purified by affinity chromatography column, and the interference on the biofunctions of CFH was further studied by the C3b binding assay and cofactor activity assay in vitro. RESULTS The prevalence of anti-CFH autoantibodies in lupus nephritis was significantly higher than that in healthy controls (8.3% (10/120) vs. 0% (0/30), P = 0.017), and no significant difference was found between the discovery and the validation group (8.3% (10/120) vs. 11.7% (7/60), P = 0.268) or the discovery and the NR-SLE group (8.3% (10/120) vs. 11.8% (4/34), P = 0.231). The subclass was mainly IgG2 (7/10), and major epitopes were in the middle (8/10 in SCRs 11-14) and N-terminal (7/10 in SCRs 1-4) regions of CFH. Patients with anti-CFH autoantibodies had a significantly lower prevalence of acute kidney injury (0% (0/10) vs. 40.0%(4/10), P = 0.025), lower serum creatinine levels (0.76 (0.40, 1.06) vs. 1.43 (0.46, 11.15), mg/dL, P = 0.023), and higher hemoglobin levels (113.8 ± 24.63 vs. 90.0 ± 22.53, g/L, P = 0.037) than those who were negative after further stratified analysis. A functional study showed that anti-CFH autoantibodies purified from patients with lupus nephritis could improve the binding between CFH and C3b, and also enhance the cofactor activity of CFH in vitro. CONCLUSIONS Anti-CFH autoantibodies were detected in patients with lupus nephritis in approximately 10% of patients with polyepitopes and IgG2 subclass predominance. Patients with anti-CFH autoantibodies presented with milder renal damage, and the purified autoantibodies could enhance the C3b binding and CFI cofactor activity of CFH in vitro, which suggested a protective role in the lupus nephritis.
Collapse
|
33
|
Zipfel PF, Wiech T, Stea ED, Skerka C. CFHR Gene Variations Provide Insights in the Pathogenesis of the Kidney Diseases Atypical Hemolytic Uremic Syndrome and C3 Glomerulopathy. J Am Soc Nephrol 2020; 31:241-256. [PMID: 31980588 PMCID: PMC7003313 DOI: 10.1681/asn.2019050515] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Sequence and copy number variations in the human CFHR-Factor H gene cluster comprising the complement genes CFHR1, CFHR2, CFHR3, CFHR4, CFHR5, and Factor H are linked to the human kidney diseases atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy. Distinct genetic and chromosomal alterations, deletions, or duplications generate hybrid or mutant CFHR genes, as well as hybrid CFHR-Factor H genes, and alter the FHR and Factor H plasma repertoire. A clear association between the genetic modifications and the pathologic outcome is emerging: CFHR1, CFHR3, and Factor H gene alterations combined with intact CFHR2, CFHR4, and CFHR5 genes are reported in atypical hemolytic uremic syndrome. But alterations in each of the five CFHR genes in the context of an intact Factor H gene are described in C3 glomerulopathy. These genetic modifications influence complement function and the interplay of the five FHR proteins with each other and with Factor H. Understanding how mutant or hybrid FHR proteins, Factor H::FHR hybrid proteins, and altered Factor H, FHR plasma profiles cause pathology is of high interest for diagnosis and therapy.
Collapse
Affiliation(s)
- Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany;
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany; and
| | - Thorsten Wiech
- Section of Nephropathology, Institute of Pathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Emma D Stea
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| |
Collapse
|
34
|
Rodríguez FJ, Rios HA, Aguilar MC, Rosenstiehl SM, Gelvez N, Lopez G, Tamayo ML. Genetic association with intravitreal ranibizumab response for neovascular age-related macular degeneration in Hispanic population. Taiwan J Ophthalmol 2020; 9:243-248. [PMID: 31942429 PMCID: PMC6947742 DOI: 10.4103/tjo.tjo_72_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 10/27/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND/PURPOSE: Age-related macular degeneration (AMD) is the leading cause of visual impairment in patients over 55 years. Currently, the most common therapies for neovascular AMD (nAMD) are intravitreal antiangiogenics. Studies suggest that genetic factors influence on antiangiogenics therapy outcomes. The purpose of this work was to establish the association between complement factor H (CFH) (Y402H), age-related maculopathy susceptibility 2 (ARMS2) (A69S), and high-temperature requirement factor A1 (HTRA1) (rs11200638) polymorphisms and the response to treatment with ranibizumab in patients with nAMD. METHODS: A cross-sectional study with 61 eyes with nAMD treated with ranibizumab was performed. Association between polymorphisms from CFH, ARMS2, and HTRA1 with the response to treatment was established. RESULTS: The mean age of patients was 76.6 (51–91) years. Only 37.7% of patients had a functional response and 26.2% had an anatomic response. TT polymorphism Y402H from CFH gene was associated with an increased likelihood of functional response to treatment. Otherwise, there was not a statistically significant association between anatomic and functional response to gene polymorphisms rs11200638 from HTRA1 and rs10490924 from ARMS 2. CONCLUSIONS: This study suggests that the response to intravitreal antiangiogenic therapy with ranibizumab was not associated to main polymorphisms from genes HTRA1 and ARMS2. However, it was found that the response to treatment differed according to CFH genotype, suggesting that further investigations are needed to establish if patients with the CC and TC genotype may need to be monitored more closely for disease recurrence than the TT genotype.
Collapse
Affiliation(s)
- Francisco Jose Rodríguez
- Fundacion Oftalmologica Nacional, Bogota, Colombia.,Universidad del Rosario, School of Medicine, Bogota, Colombia
| | - Hernan Andres Rios
- Fundacion Oftalmologica Nacional, Bogota, Colombia.,Universidad del Rosario, School of Medicine, Bogota, Colombia
| | - María Camila Aguilar
- Fundacion Oftalmologica Nacional, Bogota, Colombia.,Universidad del Rosario, School of Medicine, Bogota, Colombia
| | | | - Nancy Gelvez
- Institute of Human Genetics, Pontificia Universidad Javeriana, Bogota, Colombia
| | - Greizy Lopez
- Institute of Human Genetics, Pontificia Universidad Javeriana, Bogota, Colombia
| | - Martha L Tamayo
- Institute of Human Genetics, Pontificia Universidad Javeriana, Bogota, Colombia
| |
Collapse
|
35
|
Moreno-Torres A, Malvido-Jiménez IR, de la Peña-Moctezuma A, Castillo Sánchez LO, Fraga TR, Barbosa AS, Isaac L, Sahagún-Ruiz A. Culture-attenuated pathogenic Leptospira lose the ability to survive to complement-mediated-killing due to lower expression of factor H binding proteins. Microbes Infect 2019; 21:377-385. [DOI: 10.1016/j.micinf.2019.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 01/24/2019] [Accepted: 03/08/2019] [Indexed: 01/07/2023]
|
36
|
Jia K, Ma L, Wu S, Yang W. Serum Levels of Complement Factors C1q, Bb, and H in Normal Pregnancy and Severe Pre-Eclampsia. Med Sci Monit 2019; 25:7087-7093. [PMID: 31541546 PMCID: PMC6767947 DOI: 10.12659/msm.915777] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background This study aimed to determine the diagnostic role of serum levels of complement C1q, Bb, and H in nonpregnant women, women with normal pregnancy, and women with severe pre-eclampsia. Material/Methods Healthy nonpregnant women (n=30), women with early, middle, and late normal pregnancy (n=30, respectively), and women with severe pre-eclampsia (n=73) were studied. The pre-eclampsia study group included early-onset cases (n=43) and late-onset cases (n=30). Serum levels of Bb were determined by enzyme-linked immunosorbent assay (ELISA), and C1q and H were tested by a turbidimetric immunoassay method. Results In the pre-eclampsia study group, compared with women with normal pregnancy, serum levels of C1q remained stable throughout pregnancy, and Bb levels declined from mid-pregnancy (p=0.250). Serum levels of factor H increased in the middle and late stages of pregnancy, and C1q and H were lower in early-onset severe pre-eclampsia (p<0.001, p=0.009, respectively) and late-onset severe pre-eclampsia (p<0.001, p=0.031, respectively) compared with the early-onset control and late-onset control groups. Serum levels of Bb increased in early-onset severe pre-eclampsia (p=0.001) and late-onset severe pre-eclampsia (p=0.003) compared with early-onset control and late-onset control groups. The area under the receiver operator curve (ROC) for serum C1q, Bb, and H for the diagnosis of early-onset severe pre-eclampsia were 0.814 (95% CI, 0.712–0.917), 0.743 (95% CI, 0.638–0.859), and 0.681(95% CI, 0.556–0.806), and late-onset severe pre-eclampsia were 0.805 (95% CI, 0.694–0.913), 0.796 (95% CI, 0.680–0.911), and 0.662 (95% CI, 0.524–0.800). Conclusions The classical and alternative pathways of complement were activated in patients with severe pre-eclampsia. Serum levels of C1q, Bb, and H should be studied further as potential diagnostic markers for severe pre-eclampsia.
Collapse
Affiliation(s)
- Keke Jia
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China (mainland)
| | - Lijuan Ma
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China (mainland)
| | - Siyi Wu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China (mainland)
| | - Wang Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (mainland)
| |
Collapse
|
37
|
Valoti E, Noris M, Perna A, Rurali E, Gherardi G, Breno M, Parvanova Ilieva A, Petrov Iliev I, Bossi A, Trevisan R, Dodesini AR, Ferrari S, Stucchi N, Benigni A, Remuzzi G, Ruggenenti P. Impact of a Complement Factor H Gene Variant on Renal Dysfunction, Cardiovascular Events, and Response to ACE Inhibitor Therapy in Type 2 Diabetes. Front Genet 2019; 10:681. [PMID: 31428128 PMCID: PMC6689971 DOI: 10.3389/fgene.2019.00681] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/28/2019] [Indexed: 12/26/2022] Open
Abstract
Complement activation has been increasingly implicated in the pathogenesis of type 2 diabetes and its chronic complications. It is unknown whether complement factor H (CFH) genetic variants, which have been previously associated with complement-mediated organ damage likely due to inefficient complement modulation, influence the risk of renal and cardiovascular events and response to therapy with angiotensin-converting enzyme inhibitors (ACEi) in type 2 diabetic patients. Here, we have analyzed the c.2808G>T, (p.Glu936Asp) CFH polymorphism, which tags the H3 CFH haplotype associated to low plasma factor H levels and predisposing to atypical hemolytic uremic syndrome, in 1,158 type 2 diabetics prospectively followed in the Bergamo nephrologic complications of type 2 diabetes randomized, controlled clinical trial (BENEDICT) that evaluated the effect of the ACEi trandolapril on new onset microalbuminuria. At multivariable Cox analysis, the p.Glu936Asp polymorphism (Asp/Asp homozygotes, recessive model) was associated with increased risk of microalbuminuria [adjusted hazard ratio (HR) 3.25 (95% CI 1.46–7.24), P = 0.0038] and cardiovascular events [adjusted HR 2.68 (95% CI 1.23–5.87), P = 0.013]. The p.Glu936Asp genotype significantly interacted with ACEi therapy in predicting microalbuminuria. ACEi therapy was not nephroprotective in Asp/Asp homozygotes [adjusted HR 1.54 (0.18–13.07), P = 0.691 vs. non-ACEi-treated Asp/Asp patients], whereas it significantly reduced microalbuminuria events in Glu/Asp or Glu/Glu patients [adjusted HR 0.38 (0.24–0.60), P < 0.0001 vs. non-ACEi-treated Glu/Asp or Glu/Glu patients]. Among ACEi-treated patients, the risk of developing cardiovascular events was higher in Asp/Asp homozygotes than in Glu/Asp or Glu/Glu patients [adjusted HR 3.26 (1.29–8.28), P = 0.013]. Our results indicate that type 2 diabetic patients Asp/Asp homozygotes in the p.Glu936Asp CFH polymorphism are at increased risk of microalbuminuria and cardiovascular complications and may be less likely to benefit from ACEi therapy. Further studies are required to confirm our findings.
Collapse
Affiliation(s)
- Elisabetta Valoti
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Marina Noris
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Annalisa Perna
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Erica Rurali
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Giulia Gherardi
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Matteo Breno
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Aneliya Parvanova Ilieva
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Ilian Petrov Iliev
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Antonio Bossi
- Units of Diabetology of Treviglio Hospital, Treviglio, Italy
| | - Roberto Trevisan
- Unit of Diabetology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | | | - Silvia Ferrari
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Nadia Stucchi
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Ariela Benigni
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy
| | - Giuseppe Remuzzi
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy.,Unit of Nephrology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy.,Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Piero Ruggenenti
- Aldo e Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Ranica, Italy.,Unit of Nephrology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
38
|
Altered Peripheral Blood Leucocyte Phenotype and Responses in Healthy Individuals with Homozygous Deletion of FHR1 and FHR3 Genes. J Clin Immunol 2019; 39:336-345. [DOI: 10.1007/s10875-019-00619-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/26/2019] [Indexed: 01/10/2023]
|
39
|
Gourgari E, Ma J, Playford MP, Mehta NN, Goldman R, Remaley AT, Gordon SM. Proteomic alterations of HDL in youth with type 1 diabetes and their associations with glycemic control: a case-control study. Cardiovasc Diabetol 2019; 18:43. [PMID: 30922315 PMCID: PMC6437869 DOI: 10.1186/s12933-019-0846-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/18/2019] [Indexed: 11/12/2022] Open
Abstract
Background Patients with type 1 diabetes (T1DM) typically have normal or even elevated plasma high density lipoprotein (HDL) cholesterol concentrations; however, HDL protein composition can be altered without a change in cholesterol content. Alteration of the HDL proteome can result in dysfunctional HDL particles with reduced ability to protect against cardiovascular disease (CVD). The objective of this study was to compare the HDL proteomes of youth with T1DM and healthy controls (HC) and to evaluate the influence of glycemic control on HDL protein composition. Methods This was a cross-sectional case–control study. Blood samples were obtained from patients with T1DM and HC. HDL was isolated from plasma by size-exclusion chromatography and further purified using a lipid binding resin. The HDL proteome was analyzed by mass spectrometry using label-free SWATH peptide quantification. Results Samples from 26 patients with T1DM and 13 HC were analyzed and 78 HDL-bound proteins were measured. Youth with T1DM had significantly increased amounts of complement factor H related protein 2 (FHR2; adjusted P < 0.05), compared to HC. When patients were analyzed based on glucose control, several trends emerged. Some proteins were altered in T1DM and not influenced by glycemic control (e.g. FHR2) while others were partially or completely corrected with optimal glucose control (e.g. alpha-1-beta glycoprotein, A1BG). In a subgroup of poorly controlled T1DM patients, inter alpha trypsin inhibitor 4 (ITIH4) was dramatically elevated (P < 0.0001) and this was partially reversed in patients with optimal glucose control. Some proteins including complement component C3 (CO3) and albumin (ALB) were significantly different only in T1DM patients with optimal glucose control, suggesting a possible effect of exogenous insulin. Conclusions Youth with T1DM have proteomic alterations of their HDL compared to HC, despite similar concentration of HDL cholesterol. The influence of these compositional changes on HDL function are not yet known. Future efforts should focus on investigating the role of these HDL associated proteins in regard to HDL function and their role in CVD risk in patients with T1DM. Trial registration NCT02275091 Electronic supplementary material The online version of this article (10.1186/s12933-019-0846-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evgenia Gourgari
- Division of Pediatric Endocrinology, Department of Pediatrics, Georgetown University, Washington, DC, 20016, USA.
| | - Junfeng Ma
- Proteomics and Metabolomics Shared Resource, Georgetown University Medical Center, Washington, DC, USA.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Martin P Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Radoslav Goldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Scott M Gordon
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA.,Department of Physiology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
40
|
Top O, Parsons J, Bohlender LL, Michelfelder S, Kopp P, Busch-Steenberg C, Hoernstein SNW, Zipfel PF, Häffner K, Reski R, Decker EL. Recombinant Production of MFHR1, A Novel Synthetic Multitarget Complement Inhibitor, in Moss Bioreactors. FRONTIERS IN PLANT SCIENCE 2019; 10:260. [PMID: 30949184 PMCID: PMC6436476 DOI: 10.3389/fpls.2019.00260] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/19/2019] [Indexed: 05/23/2023]
Abstract
The human complement system is an important part of the immune system responsible for lysis and elimination of invading microorganisms and apoptotic body cells. Improper activation of the system due to deficiency, mutations, or autoantibodies of complement regulators, mainly factor H (FH) and FH-related proteins (FHRs), causes severe kidney and eye diseases. However, there is no recombinant FH therapeutic available on the market. The first successful recombinant production of FH was accomplished with the moss bioreactor, Physcomitrella patens. Recently, a synthetic regulator, MFHR1, was designed to generate a multitarget complement inhibitor that combines the activities of FH and the FH-related protein 1 (FHR1). The potential of MFHR1 was demonstrated in a proof-of-concept study with transiently transfected insect cells. Here, we present the stable production of recombinant glyco-engineered MFHR1 in the moss bioreactor. The key features of this system are precise genome engineering via homologous recombination, Good Manufacturing Practice-compliant production in photobioreactors, high batch-to-batch reproducibility, and product stability. Several potential biopharmaceuticals are being produced in this system. In some cases, these are even biobetters, i.e., the recombinant proteins produced in moss have a superior quality compared to their counterparts from mammalian systems as for example moss-made aGal, which successfully passed phase I clinical trials. Via mass spectrometry-based analysis of moss-produced MFHR1, we now prove the correct synthesis and modification of this glycoprotein with predominantly complex-type N-glycan attachment. Moss-produced MFHR1 exhibits cofactor and decay acceleration activities comparable to FH, and its mechanism of action on multiple levels within the alternative pathway of complement activation led to a strong inhibitory activity on the whole alternative pathway, which was higher than with the physiological regulator FH.
Collapse
Affiliation(s)
- Oguz Top
- Department of Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Juliana Parsons
- Department of Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lennard L. Bohlender
- Department of Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stefan Michelfelder
- Faculty of Medicine, Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Center - University Freiburg, University of Freiburg, Freiburg, Germany
| | - Phillipp Kopp
- Department of Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | | | | - Peter F. Zipfel
- Leibniz Institute for Natural Product Research and Infection Biology, Friedrich Schiller University, Jena, Germany
| | - Karsten Häffner
- Faculty of Medicine, Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Center - University Freiburg, University of Freiburg, Freiburg, Germany
| | - Ralf Reski
- Department of Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Eva L. Decker
- Department of Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
41
|
Song D, Mohammed I, Bhuyan R, Miwa T, Williams AL, Gullipalli D, Sato S, Song Y, Dunaief JL, Song WC. Retinal Basal Laminar Deposits in Complement fH/fP Mouse Model of Dense Deposit Disease. Invest Ophthalmol Vis Sci 2019; 59:3405-3415. [PMID: 30025090 PMCID: PMC6040236 DOI: 10.1167/iovs.18-24133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose Dense deposit disease (DDD) is caused by dysregulation of the alternative pathway of the complement cascade and characterized by electron-dense deposits in the kidney glomerular basement membrane (GBM) and drusen in Bruch's membrane (BrM). Complement factor H (fH) and factor properdin (fP) regulate complement activation; fH inhibits alternative pathway (AP) activation, whereas fP promotes it. We report pathologic changes in eyes of an fH and fP double-mutant mouse, which we previously showed have dense deposits in the GBM and early mortality from nephropathy. Methods fHm/m, fP−/−, and fHm/m/fP−/− mice were generated on a C57BL/6–129J background. Fundus imaging at 8 weeks of age was followed by analysis via light and electron microscopy. Retinal function was assessed by electroretinography (ERG). Complement levels and localization were tested by immunohistochemistry and ELISA. Retinas of fHm/m/fP−/− mice treated with intraperitoneal injections of an anti-C5 antibody were compared to those of age- and genotype-matched mice injected with an isotype control antibody. Results fHm/m/fP−/− mice suffered early-onset retinal hypopigmented spots detected using in vivo retinal photography, and histologic examination showed basal laminar deposits (BLamD), degeneration of the photoreceptors, and RPE vacuolization. ERG showed diminished retinal function. The anti-C5 antibody was retina-protective. Conclusions This unique mouse represents a new model of complement-mediated rapid-onset DDD, and could be useful in exploring the pathologic changes associated with BLamD in age-related macular degeneration.
Collapse
Affiliation(s)
- Delu Song
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Imran Mohammed
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Rupak Bhuyan
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Allison Lesher Williams
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Sayaka Sato
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Ying Song
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Joshua L Dunaief
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
42
|
Song D, Ueda Y, Bhuyan R, Mohammed I, Miwa T, Gullipali D, Kim H, Zhou L, Song Y, Schultz H, Bargoud A, Dunaief JL, Song WC. Complement Factor H Mutation W1206R Causes Retinal Thrombosis and Ischemic Retinopathy in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:826-838. [PMID: 30711487 DOI: 10.1016/j.ajpath.2019.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 12/06/2018] [Accepted: 01/02/2019] [Indexed: 01/14/2023]
Abstract
Single-nucleotide polymorphisms and rare mutations in factor H (FH; official name, CFH) are associated with age-related macular degeneration and atypical hemolytic uremic syndrome, a form of thrombotic microangiopathy. Mice with the FH W1206R mutation (FHR/R) share features with human atypical hemolytic uremic syndrome. Herein, we report that FHR/R mice exhibited retinal vascular occlusion and ischemia. Retinal fluorescein angiography demonstrated delayed perfusion and vascular leakage in FHR/R mice. Optical coherence tomography imaging of FHR/R mice showed retinal degeneration, edema, and detachment. Histologic analysis of FHR/R mice revealed retinal thinning, vessel occlusion, as well as degeneration of photoreceptors and retinal pigment epithelium. Immunofluorescence showed albumin leakage from blood vessels into the neural retina, and electron microscopy demonstrated vascular endothelial cell irregularity with narrowing of retinal and choroidal vessels. Knockout of C6, a component of the membrane attack complex, prevented the aforementioned retinal phenotype in FHR/R mice, consistent with membrane attack complex-mediated pathogenesis. Pharmacologic blockade of C5 also rescued retinas of FHR/R mice. This FHR/R mouse strain represents a model for retinal vascular occlusive disorders and ischemic retinopathy. The results suggest complement dysregulation can contribute to retinal vascular occlusion and that an anti-C5 antibody might be helpful for C5-mediated thrombotic retinal diseases.
Collapse
Affiliation(s)
- Delu Song
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yoshiyasu Ueda
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rupak Bhuyan
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Imran Mohammed
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Takashi Miwa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Damodar Gullipali
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hangsoo Kim
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lin Zhou
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ying Song
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hannah Schultz
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Albert Bargoud
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua L Dunaief
- Department of Ophthalmology, Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
43
|
Michael M, Turner N, Elenberg E, Shaffer LG, Teruya J, Arar M, Hui SK, Smith RJ, Moake J. Deficiency of complement factor H-related proteins and autoantibody-positive hemolytic uremic syndrome in an infant with combined partial deficiencies and autoantibodies to complement factor H and ADAMTS13. Clin Kidney J 2018; 11:791-796. [PMID: 30524124 PMCID: PMC6275444 DOI: 10.1093/ckj/sfy010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/23/2018] [Indexed: 11/23/2022] Open
Abstract
A 3-month-old male infant developed an extremely severe episode of atypical hemolytic uremic syndrome (aHUS) associated with partial deficiencies of full-length complement factor H (FH; ∼15% of infant normal) and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13) (39% of normal) and autoantibodies reactive with both proteins. His FH and ADAMTS13 genes were normal, indicating that the partial deficiencies were acquired, probably as the result of autoantibodies against full-length FH and ADAMTS13. The child also had a homozygous deletion of the complement factor H–related (CFHR)3–CFHR1 portion in the complement factor H (CFH) gene cluster. He therefore had deficiency of CFHR proteins and autoantibody-positive hemolytic uremic syndrome (DEAP-HUS) with an unusual early onset associated with a partial deficiency of ADAMTS13 and an anti-ADAMTS13 autoantibody. His clinical episode of aHUS responded to plasma infusion and subsequent treatment with mycophenolate and rituximab. We believe that this is the first report of DEAP-HUS in an infant with partial deficiencies in both ADAMTS13 and full-length FH acquired in association with autoantibodies to both proteins.
Collapse
Affiliation(s)
- Mini Michael
- Texas Children's Hospital, Houston, TX, USA.,Baylor College of Medicine, Houston, TX, USA
| | | | - Ewa Elenberg
- Texas Children's Hospital, Houston, TX, USA.,Baylor College of Medicine, Houston, TX, USA
| | | | - Jun Teruya
- Texas Children's Hospital, Houston, TX, USA.,Baylor College of Medicine, Houston, TX, USA
| | - Mazen Arar
- University of Texas Health Science Center, San Antonio, TX, USA
| | - Shiu-Ki Hui
- Texas Children's Hospital, Houston, TX, USA.,Baylor College of Medicine, Houston, TX, USA
| | - Richard J Smith
- Molecular Otolaryngology and Renal Research Laboratories, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Joel Moake
- Baylor College of Medicine, Houston, TX, USA.,Rice University, Houston, TX, USA
| |
Collapse
|
44
|
Wijeratne SS, Nolasco L, Li J, Jiang K, Moake JL, Kiang CH. Correlating Conformational Dynamics with the Von Willebrand Factor Reductase Activity of Factor H Using Single Molecule Force Measurements. J Phys Chem B 2018; 122:10653-10658. [PMID: 30351116 DOI: 10.1021/acs.jpcb.8b06153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Activation of proteins often involves conformational transitions, and these switches are often difficult to characterize in multidomain proteins. Full-length factor H (FH), consisting of 20 small consensus repeat domains (150 kD), is a complement control protein that regulates the activity of the alternative complement pathway. Different preparations of FH can also reduce the disulfide bonds linking large Von Willebrand factor (VWF) multimers into smaller, less adhesive forms. In contrast, commercially available purified FH (pFH) has little or no VWF reductase activity unless the pFH is chemically modified by either ethylenediaminetetraacetic acid (EDTA) or urea. We used atomic force microscopy single molecule force measurements to investigate different forms of FH, including recombinant FH and pFH, in the presence or absence of EDTA and urea, and to correlate the conformational changes to its activities. We found that the FH conformation depends on the method used for sample preparation, which affects the VWF reductase activity of FH.
Collapse
|
45
|
Maugeri A, Barchitta M, Mazzone MG, Giuliano F, Agodi A. Complement System and Age-Related Macular Degeneration: Implications of Gene-Environment Interaction for Preventive and Personalized Medicine. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7532507. [PMID: 30225264 PMCID: PMC6129329 DOI: 10.1155/2018/7532507] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/18/2018] [Indexed: 12/14/2022]
Abstract
Age-related macular degeneration (AMD) is the most common cause of visual loss in developed countries, with a significant economic and social burden on public health. Although genome-wide and gene-candidate studies have been enabled to identify genetic variants in the complement system associated with AMD pathogenesis, the effect of gene-environment interaction is still under debate. In this review we provide an overview of the role of complement system and its genetic variants in AMD, summarizing the consequences of the interaction between genetic and environmental risk factors on AMD onset, progression, and therapeutic response. Finally, we discuss the perspectives of current evidence in the field of genomics driven personalized medicine and public health.
Collapse
Affiliation(s)
- Andrea Maugeri
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
| | - Martina Barchitta
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
| | - Maria Grazia Mazzone
- SIFI SpA, Research and Development Department, Via Ercole Patti 36, 95025 Catania, Italy
| | - Francesco Giuliano
- SIFI SpA, Research and Development Department, Via Ercole Patti 36, 95025 Catania, Italy
| | - Antonella Agodi
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
| |
Collapse
|
46
|
Ho EXP, Cheung CMG, Sim S, Chu CW, Wilm A, Lin CB, Mathur R, Wong D, Chan CM, Bhagarva M, Laude A, Lim TH, Wong TY, Cheng CY, Davila S, Hibberd M. Human pharyngeal microbiota in age-related macular degeneration. PLoS One 2018; 13:e0201768. [PMID: 30089174 PMCID: PMC6082546 DOI: 10.1371/journal.pone.0201768] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/20/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND While the aetiology of age-related macular degeneration (AMD)-a major blinding disease-remains unknown, the disease is strongly associated with variants in the complement factor H (CFH) gene. CFH variants also confer susceptibility to invasive infection with several bacterial colonizers of the nasopharyngeal mucosa. This shared susceptibility locus implicates complement deregulation as a common disease mechanism, and suggests the possibility that microbial interactions with host complement may trigger AMD. In this study, we address this possibility by testing the hypothesis that AMD is associated with specific microbial colonization of the human nasopharynx. RESULTS High-throughput Illumina sequencing of the V3-V6 region of the microbial 16S ribosomal RNA gene was used to comprehensively and accurately describe the human pharyngeal microbiome, at genus level, in 245 AMD patients and 386 controls. Based on mean and differential microbial abundance analyses, we determined an overview of the pharyngeal microbiota, as well as candidate genera (Prevotella and Gemella) suggesting an association towards AMD health and disease conditions. CONCLUSIONS Utilizing an extensive study population from Singapore, our results provided an accurate description of the pharyngeal microbiota profiles in AMD health and disease conditions. Through identification of candidate genera that are different between conditions, we provide preliminary evidence for the existence of microbial triggers for AMD. Ethical approval for this study was obtained through the Singapore Health Clinical Institutional Review Board, reference numbers R799/63/2010 and 2010/585/A.
Collapse
Affiliation(s)
| | - Chui Ming Gemmy Cheung
- Singapore Eye Research Institute, Singapore National Eye Center, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Shuzhen Sim
- Genome Institute of Singapore,Singapore, Singapore
| | | | - Andreas Wilm
- Genome Institute of Singapore,Singapore, Singapore
| | | | - Ranjana Mathur
- Singapore Eye Research Institute, Singapore National Eye Center, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Doric Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Choi Mun Chan
- Singapore Eye Research Institute, Singapore National Eye Center, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Mayuri Bhagarva
- Department of Ophthalmology, National University of Singapore and National University Health System, Singapore, Singapore
| | - Augustinus Laude
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tock Han Lim
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Ching Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Center, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Sonia Davila
- Genome Institute of Singapore,Singapore, Singapore
| | - Martin Hibberd
- Genome Institute of Singapore,Singapore, Singapore
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
47
|
Qi P, Wu B, Guo B, Zhang C, Xu K. The complement factor H (CFH) and its related protein 2 (CFHR2) mediating immune response in large yellow croaker Larimichthys crocea. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 84:241-249. [PMID: 29496498 DOI: 10.1016/j.dci.2018.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/25/2018] [Accepted: 02/25/2018] [Indexed: 06/08/2023]
Abstract
Complement is a complex innate immune surveillance system, playing a key role in host homeostasis, inflammation, and in the defense against pathogens. Complement regulators are crucial to prevent the injudicious production of these mediators and potential injury to self tissues. Here, we identified the complement factor H (CFH) and its related gene 2 (CFHR2) homologs from large yellow croaker (Larimichthys crocea), named LcCfh and LcCfhr2, respectively. The deduced LcCfh and LcCfhr2 proteins shared significant structural similarities and identified codes for a polypeptide consisting of various numbers of highly conserved SCR domains. LcCfh, LcCfhr1 and LcCfhr2 genes were detected in all examined tissues with predominantly expressions in liver, spleen and kidney, and their expressions all increased upon Vibrio alginolyticus challenge. In vitro assays showed that recombinant LcCfh was likely to act as a cofactor of CFI and played a negative regulation role in complement system, when recombinant LcCfhr2 seemed to play mechanisms independent of the activity of CFH. Both recombinant LcCfh and LcCfhr2 took participate in inflammatory reaction despite of the inequal ability to mediate pro-inflammation response. These data provide a new insight into the functional activities of teleost complement system.
Collapse
Affiliation(s)
- Pengzhi Qi
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, 316004, China.
| | - Bin Wu
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, 316004, China
| | - Baoying Guo
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, 316004, China
| | - Chuan Zhang
- Zhoushan Aquatic Science Research Institute, Zhoushan, 316000, China
| | - Kaida Xu
- Scientific Observing and Experimental Station of Fishery Resources for Key Fishing Grounds, MOA, Key Laboratory of Sustainable Utilization of Technology Research, Marine Fisheries Research Institute of Zhejiang, Zhoushan, 316021, China
| |
Collapse
|
48
|
Goicoechea de Jorge E, López Lera A, Bayarri-Olmos R, Yebenes H, Lopez-Trascasa M, Rodríguez de Córdoba S. Common and rare genetic variants of complement components in human disease. Mol Immunol 2018; 102:42-57. [PMID: 29914697 DOI: 10.1016/j.molimm.2018.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/06/2018] [Accepted: 06/08/2018] [Indexed: 12/21/2022]
Abstract
Genetic variability in the complement system and its association with disease has been known for more than 50 years, but only during the last decade have we begun to understand how this complement genetic variability contributes to the development of diseases. A number of reports have described important genotype-phenotype correlations that associate particular diseases with genetic variants altering specific aspects of the activation and regulation of the complement system. The detailed functional characterization of some of these genetic variants provided key insights into the pathogenic mechanisms underlying these pathologies, which is facilitating the design of specific anti-complement therapies. Importantly, these analyses have sometimes revealed unknown features of the complement proteins. As a whole, these advances have delineated the functional implications of genetic variability in the complement system, which supports the implementation of a precision medicine approach based on the complement genetic makeup of the patients. Here we provide an overview of rare complement variants and common polymorphisms associated with disease and discuss what we have learned from them.
Collapse
Affiliation(s)
- Elena Goicoechea de Jorge
- Department of Immunology, Complutense University, Madrid, Spain; Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alberto López Lera
- Research Institute Hospital Universitario La Paz (IdiPaz), Madrid, Spain; Ciber de Enfermedades Raras, Madrid, Spain
| | - Rafael Bayarri-Olmos
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Hugo Yebenes
- Ciber de Enfermedades Raras, Madrid, Spain; Molecular Pathology and Complement Genetics Unit. Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | | - Santiago Rodríguez de Córdoba
- Ciber de Enfermedades Raras, Madrid, Spain; Molecular Pathology and Complement Genetics Unit. Centro de Investigaciones Biológicas, CSIC, Madrid, Spain.
| |
Collapse
|
49
|
Lidani KCF, Sandri TL, Andrade FA, Bavia L, Nisihara R, Messias-Reason IJ. Complement Factor H as a potential atherogenic marker in chronic Chagas’ disease. Parasite Immunol 2018; 40:e12537. [DOI: 10.1111/pim.12537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/10/2018] [Indexed: 02/03/2023]
Affiliation(s)
- K. C. F. Lidani
- Laboratory of Molecular Immunopathology; Clinical Hospital; Federal University of Paraná; Curitiba Brazil
| | - T. L. Sandri
- Laboratory of Molecular Immunopathology; Clinical Hospital; Federal University of Paraná; Curitiba Brazil
- Institute of Tropical Medicine; University of Tübingen; Tübingen Germany
| | - F. A. Andrade
- Laboratory of Molecular Immunopathology; Clinical Hospital; Federal University of Paraná; Curitiba Brazil
| | - L. Bavia
- Laboratory of Molecular Immunopathology; Clinical Hospital; Federal University of Paraná; Curitiba Brazil
| | - R. Nisihara
- Laboratory of Molecular Immunopathology; Clinical Hospital; Federal University of Paraná; Curitiba Brazil
| | - I. J. Messias-Reason
- Laboratory of Molecular Immunopathology; Clinical Hospital; Federal University of Paraná; Curitiba Brazil
| |
Collapse
|
50
|
Recurrent structural variation, clustered sites of selection, and disease risk for the complement factor H ( CFH) gene family. Proc Natl Acad Sci U S A 2018; 115:E4433-E4442. [PMID: 29686068 DOI: 10.1073/pnas.1717600115] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Structural variation and single-nucleotide variation of the complement factor H (CFH) gene family underlie several complex genetic diseases, including age-related macular degeneration (AMD) and atypical hemolytic uremic syndrome (AHUS). To understand its diversity and evolution, we performed high-quality sequencing of this ∼360-kbp locus in six primate lineages, including multiple human haplotypes. Comparative sequence analyses reveal two distinct periods of gene duplication leading to the emergence of four CFH-related (CFHR) gene paralogs (CFHR2 and CFHR4 ∼25-35 Mya and CFHR1 and CFHR3 ∼7-13 Mya). Remarkably, all evolutionary breakpoints share a common ∼4.8-kbp segment corresponding to an ancestral CFHR gene promoter that has expanded independently throughout primate evolution. This segment is recurrently reused and juxtaposed with a donor duplication containing exons 8 and 9 from ancestral CFH, creating four CFHR fusion genes that include lineage-specific members of the gene family. Combined analysis of >5,000 AMD cases and controls identifies a significant burden of a rare missense mutation that clusters at the N terminus of CFH [P = 5.81 × 10-8, odds ratio (OR) = 9.8 (3.67-Infinity)]. A bipolar clustering pattern of rare nonsynonymous mutations in patients with AMD (P < 10-3) and AHUS (P = 0.0079) maps to functional domains that show evidence of positive selection during primate evolution. Our structural variation analysis in >2,400 individuals reveals five recurrent rearrangement breakpoints that show variable frequency among AMD cases and controls. These data suggest a dynamic and recurrent pattern of mutation critical to the emergence of new CFHR genes but also in the predisposition to complex human genetic disease phenotypes.
Collapse
|