1
|
Pan Q, Huang X, Liu C, Pan Q, Huang S. Systemic lupus erythematosus and atherosclerosis: immune pathways and the uncharted territory of gut microbiota and metabolism. Front Immunol 2025; 16:1492726. [PMID: 40356907 PMCID: PMC12067219 DOI: 10.3389/fimmu.2025.1492726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Patients with Systemic Lupus Erythematosus (SLE) are significantly more susceptible to atherosclerosis, which may elevate their mortality risk. The review explores recent understandings of the origins and remedies for atherosclerosis associated with SLE. Our focus is particularly on the consequences of immune system disparities, interruptions in intestinal bacteria, and metabolic complications. The influence of SLE on atherosclerosis extends past usual risk elements, including processes specific to the disease. The list encompasses excessive immune cell activity, production of autoantibodies, inflammatory responses. A variety of therapies for atherosclerosis linked to SLE encompass cholesterol-lowering medications, anti-inflammatory drugs, immune suppressors, antimalarials, interferon treatments, NET inhibitors, and methods aimed at T and B-cells. However, existing research has its shortcomings, necessitating additional clinical trials to ascertain the efficacy and security of these therapies. The direct interactions among SLE, gut microbiota, metabolism, and atherosclerosis is underexplored, presenting innovation opportunities. Research into specific gut microbial strains and metabolites' effects on immune responses and atherosclerosis progression in SLE patients is needed. Such research could uncover novel therapeutic targets and biomarkers, advancing prevention and treatment strategies for SLE cardiovascular complications.
Collapse
Affiliation(s)
- Quanren Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xuemei Huang
- Laboratory of Cardiovascular Diseases, Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chaobin Liu
- Laboratory of Cardiovascular Diseases, Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shian Huang
- Laboratory of Cardiovascular Diseases, Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
2
|
Orefice V, Ceccarelli F, Barbati C, Buoncuore G, Pirone C, Alessandri C, Conti F. Caffeine improves systemic lupus erythematosus endothelial dysfunction by promoting endothelial progenitor cells survival. Rheumatology (Oxford) 2025; 64:1886-1893. [PMID: 39380132 DOI: 10.1093/rheumatology/keae453] [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: 11/19/2023] [Accepted: 06/20/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVE We studied the role of caffeine intake on endothelial function in SLE by assessing its effect on circulating endothelial progenitor cells (EPCs) both ex vivo in SLE patients and in vitro in healthy donors (HD) treated with SLE sera. METHODS We enrolled SLE patients without traditional cardiovascular risks factors. Caffeine intake was evaluated with a 7-day food frequency questionnaire. EPCs percentage was assessed by flow cytometry analysis and, subsequently, EPCs pooled from six HD were co-cultured with caffeine with and without SLE sera. After 7 days, we evaluated cells' morphology and ability to form colonies, the percentage of apoptotic cells by flow cytometry analysis and the levels of autophagy and apoptotic markers by western blot. Finally, we performed a western blot analysis to assess the A2AR/SIRT3/AMPK pathway. RESULTS We enrolled 31 SLE patients, and observed a positive correlation between caffeine intake and circulating EPCs percentage. HD EPCs treated with SLE sera and caffeine showed an improvement in morphology and in number of EPCs colony-forming units in comparison with those incubated without caffeine. Caffeine was able to restore autophagy and apoptotic markers in HD EPCs as before SLE sera treatment. Finally, caffeine treatment was able to significantly reduce A2AR levels, leading to an increase in protein levels of SIRT3 and subsequently AMPK phosphorylation. CONCLUSIONS Caffeine intake positively correlated with the percentage of circulating EPCs in SLE patients; moreover, caffeine in vitro treatment was able to improve EPC survival and vitality through the inhibition of apoptosis and the promotion of autophagy via A2AR/SIRT3/AMPK pathway.
Collapse
Affiliation(s)
- Valeria Orefice
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Fulvia Ceccarelli
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Cristiana Barbati
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Giorgia Buoncuore
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Carmelo Pirone
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
3
|
Whittall Garcia LP, Gladman DD, Urowitz M, Bonilla D, Schneider R, Touma Z, Wither J. Interferon-α as a biomarker to predict renal outcomes in lupus nephritis. Lupus Sci Med 2024; 11:e001347. [PMID: 39613334 DOI: 10.1136/lupus-2024-001347] [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: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 12/01/2024]
Abstract
OBJECTIVE To determine if serum interferon (IFN)-α levels at the time of a lupus nephritis (LN) flare are associated with renal outcomes. METHODS Patients with an LN flare who had a preflare estimated glomerular filtration rate (eGFR) ≥60 mL/min were included in the study. The following outcomes were ascertained: (1) Time to first and second LN flares during follow-up, (2) Time to a sustained decline in eGFR by 30% and 50%, and progression to end-stage renal disease (ESRD, <15 mL/min), and (3) Time to an adverse renal event (≥2 renal flares and/or at least a 30% sustained decline in eGFR during follow-up). Serum IFN-α was measured by Simoa. RESULTS 92 patients with active LN were included in the study. Elevated serum baseline levels of IFN-α predicted poor renal outcomes. Patients with higher baseline IFN-α had a greater risk of having two or more subsequent LN flares (HR: 1.31 (1.08-1.59), p=0.006), sustained 30% decline in eGFR (HR: 1.27 (1.14-1.40), p<0.001), 50% decline in eGFR (HR: 1.27 (1.12-1.33), p<0.001) and progressing to ESRD (HR: 1.29 (1.14-1.47), p<0.001). Receiver operating characteristic analysis identified an IFN-α cut-off, 0.6 pg/ml, for predicting an adverse renal event. CONCLUSIONS Elevated serum IFN-α levels measured at the time of an LN flare are associated with poor renal outcomes, including the development of ≥2 LN flares, and a clinically meaningful decline in kidney function.
Collapse
Affiliation(s)
| | - Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Murray Urowitz
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Dennisse Bonilla
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Raphael Schneider
- Department of Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Zahi Touma
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Joan Wither
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Mangoni AA, Zinellu A. The vascular endothelial growth factor as a candidate biomarker of systemic lupus erythematosus: a GRADE-assessed systematic review and meta-analysis. Clin Exp Med 2024; 24:218. [PMID: 39259392 PMCID: PMC11390800 DOI: 10.1007/s10238-024-01487-w] [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: 06/26/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
There is an ongoing search for novel biomarkers of endothelial damage, active disease, and organ dysfunction in systemic lupus erythematosus (SLE). We investigated the role of the vascular endothelial growth factor (VEGF) as a candidate biomarker by conducting a systematic review and meta-analysis of studies examining VEGF concentrations in SLE patients and healthy controls. We searched electronic databases (PubMed, Scopus, and Web of Science) from inception to 31 May 2024 (inclusion criteria: VEGF measurement in SLE patients and healthy controls and SLE patients with and without active disease or specific organ dysfunction in case-control studies, recruitment of adult participants, and availability of the full text in the English language; exclusion criteria: non-case-control studies, participants under 18 years, articles reporting duplicate or irrelevant data, and animal studies). We assessed the risk of bias and the certainty of evidence using the JBI Critical Appraisal Checklist and GRADE, respectively (PROSPERO registration number: CRD42024561636). Circulating VEGF concentrations were significantly higher in SLE patients than in controls (22 studies; standardised mean difference, SMD = 0.71, 95% CI 0.44 to 0.98, p < 0.001; low certainty of evidence). In SLE patients, VEGF concentrations were significantly higher in those with active disease (six studies; SMD = 1.10, 95% CI 0.27 to 1.92, p = 0.009; very low certainty of evidence) and lupus nephritis (four studies; SMD = 0.80, 95% CI 0.03 to 1.57, p = 0.042; very low certainty of evidence). Only one study reported VEGF concentrations in SLE patients with and without pulmonary arterial hypertension. The effect size of the differences in VEGF concentrations between SLE patients and controls was not associated with disease duration, use of glucocorticoids and immunosuppressors, biological matrix assessed, or analytical method used. However, it was significantly associated with the study's geographical location. The evidence was limited by the high but partially explainable heterogeneity and the presence of publication bias which was addressed with the "trim-and-fill" method (SLE presence), the high but partially explainable heterogeneity and lack of assessment of publication bias because of the limited study number (active disease), and the limited study number preventing the identification of sources of heterogeneity, sensitivity analysis, and assessment of publication bias (lupus nephritis). Our results highlight VEGF's potential role as a SLE biomarker and the need for further research, also given the aforementioned limitations, investigating VEGF concentrations in a wide range of SLE patient subgroups.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, SA, 5042, Australia.
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
5
|
Zhang Y, Liu K, Guo M, Yang Y, Zhang H. Negative regulator IL-1 receptor 2 (IL-1R2) and its roles in immune regulation of autoimmune diseases. Int Immunopharmacol 2024; 136:112400. [PMID: 38850793 DOI: 10.1016/j.intimp.2024.112400] [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: 04/09/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
The decoy receptor interleukin 1 receptor 2 (IL-1R2), also known as CD121b, has different forms: membrane-bound (mIL-1R2), soluble secreted (ssIL-1R2), shedded (shIL-1R2), intracellular domain (IL-1R2ICD). The different forms of IL-1R2 exert not exactly similar functions. IL-1R2 can not only participate in the regulation of inflammatory response by competing with IL-1R1 to bind IL-1 and IL-1RAP, but also regulate IL-1 maturation and cell activation, promote cell survival, participate in IL-1-dependent internalization, and even have biological activity as a transcriptional cofactor. In this review, we provide a detailed description of the biological characteristics of IL-1R2 and discuss the expression and unique role of IL-1R2 in different immune cells. Importantly, we summarize the role of IL-1R2 in immune regulation from different autoimmune diseases, hoping to provide a new direction for in-depth studies of pathogenesis and therapeutic targets in autoimmune diseases.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha City, Hunan Province, China
| | - Yiying Yang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China; Postdoctoral Research Station of Biology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China.
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
6
|
Visser A, van Nimwegen JF, Wilbrink R, Liefers SC, van der Tuuk K, Mourits MJE, Diercks GFH, Bart J, van der Vegt B, van Kempen LC, Bootsma H, Kroese FGM, Verstappen GM. Increased Interferon Signaling in Vaginal Tissue of Patients With Primary Sjögren Syndrome. J Rheumatol 2024; 51:687-695. [PMID: 38561184 DOI: 10.3899/jrheum.2023-1068] [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] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Vaginal dryness is an important factor influencing sexual function in women with primary Sjögren syndrome (pSS). Previous studies showed a higher degree of inflammation in vaginal biopsies from patients with pSS compared to non-pSS controls. However, the molecular pathways that drive this inflammation remain unclear. Therefore, the aim of this study was to investigate inflammatory pathway activity in the vaginal tissue of patients with pSS. METHODS Vaginal biopsies of 8 premenopausal patients with pSS experiencing vaginal dryness and 7 age-matched non-pSS controls were included. Expression of genes involved in inflammation and tissue homeostasis was measured using NanoString technology and validated using TaqMan Real-Time PCR. Vaginal tissue sections were stained by immunohistochemistry for myxovirus resistance protein 1 (MxA) and CD123 (plasmacytoid dendritic cells [pDCs]). RESULTS The most enriched pathway in vaginal biopsies from patients with pSS compared to non-pSS controls was the interferon (IFN) signaling pathway (P < 0.01). Pathway scores for Janus kinase and signal transducer and activator of transcription (JAK-STAT) and Notch signaling were also higher (P < 0.01 for both pathways). Conversely, transforming growth factor-β signaling and angiogenesis pathway scores were lower in pSS (P = 0.02 and P = 0.04, respectively). Differences in IFN signaling between patients with pSS and non-pSS controls were confirmed by PCR and MxA tissue staining. No CD123+ pDCs were detected in vaginal biopsies. IFN-stimulated gene expression levels correlated positively with CD45+ cell numbers in vaginal biopsies and serum anti-SSA/Ro positivity. CONCLUSION Upregulation of IFN signaling in vaginal tissue of women with pSS, along with its association with tissue pathology, suggests that IFNs contribute to inflammation of the vaginal wall and potentially also to clinical symptomatology (ie, vaginal dryness).
Collapse
Affiliation(s)
- Annie Visser
- A. Visser, BSc, J.F. van Nimwegen, MD, PhD, R. Wilbrink, MD, S.C. Liefers, PhD, H. Bootsma, MD, PhD, F.G.M. Kroese, PhD, G.M. Verstappen, PharmD, PhD, University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Jolien F van Nimwegen
- A. Visser, BSc, J.F. van Nimwegen, MD, PhD, R. Wilbrink, MD, S.C. Liefers, PhD, H. Bootsma, MD, PhD, F.G.M. Kroese, PhD, G.M. Verstappen, PharmD, PhD, University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Rick Wilbrink
- A. Visser, BSc, J.F. van Nimwegen, MD, PhD, R. Wilbrink, MD, S.C. Liefers, PhD, H. Bootsma, MD, PhD, F.G.M. Kroese, PhD, G.M. Verstappen, PharmD, PhD, University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Silvia C Liefers
- A. Visser, BSc, J.F. van Nimwegen, MD, PhD, R. Wilbrink, MD, S.C. Liefers, PhD, H. Bootsma, MD, PhD, F.G.M. Kroese, PhD, G.M. Verstappen, PharmD, PhD, University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Karin van der Tuuk
- K. van der Tuuk, MD, PhD, M.J.E. Mourits, MD, PhD, University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynaecology, Groningen, the Netherlands
| | - Marian J E Mourits
- K. van der Tuuk, MD, PhD, M.J.E. Mourits, MD, PhD, University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynaecology, Groningen, the Netherlands
| | - Gilles F H Diercks
- G.F.H. Diercks, MD, PhD, J. Bart, MD, PhD, B. van der Vegt, MD, PhD, University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Joost Bart
- G.F.H. Diercks, MD, PhD, J. Bart, MD, PhD, B. van der Vegt, MD, PhD, University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Bert van der Vegt
- G.F.H. Diercks, MD, PhD, J. Bart, MD, PhD, B. van der Vegt, MD, PhD, University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Léon C van Kempen
- L.C. van Kempen, MD, PhD, University of Antwerp, Antwerp University Hospital, Department of Pathology, Antwerp, Belgium
| | - Hendrika Bootsma
- A. Visser, BSc, J.F. van Nimwegen, MD, PhD, R. Wilbrink, MD, S.C. Liefers, PhD, H. Bootsma, MD, PhD, F.G.M. Kroese, PhD, G.M. Verstappen, PharmD, PhD, University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Frans G M Kroese
- A. Visser, BSc, J.F. van Nimwegen, MD, PhD, R. Wilbrink, MD, S.C. Liefers, PhD, H. Bootsma, MD, PhD, F.G.M. Kroese, PhD, G.M. Verstappen, PharmD, PhD, University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Gwenny M Verstappen
- A. Visser, BSc, J.F. van Nimwegen, MD, PhD, R. Wilbrink, MD, S.C. Liefers, PhD, H. Bootsma, MD, PhD, F.G.M. Kroese, PhD, G.M. Verstappen, PharmD, PhD, University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands;
| |
Collapse
|
7
|
Abstract
Vascular disease is a major cause of morbidity and mortality in patients with systemic autoimmune diseases, particularly systemic lupus erythematosus (SLE). Although comorbid cardiovascular risk factors are frequently present in patients with SLE, they do not explain the high burden of premature vascular disease. Profound innate and adaptive immune dysregulation seems to be the primary driver of accelerated vascular damage in SLE. In particular, evidence suggests that dysregulation of type 1 interferon (IFN-I) and aberrant neutrophils have key roles in the pathogenesis of vascular damage. IFN-I promotes endothelial dysfunction directly via effects on endothelial cells and indirectly via priming of immune cells that contribute to vascular damage. SLE neutrophils are vasculopathic in part because of their increased ability to form immunostimulatory neutrophil extracellular traps. Despite improvements in clinical care, cardiovascular disease remains the leading cause of mortality among patients with SLE, and treatments that improve vascular outcomes are urgently needed. Improved understanding of the mechanisms of vascular injury in inflammatory conditions such as SLE could also have implications for common cardiovascular diseases, such as atherosclerosis and hypertension, and may ultimately lead to personalized therapeutic approaches to the prevention and treatment of this potentially fatal complication.
Collapse
Affiliation(s)
- William G Ambler
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
8
|
Hu H, Wu A, Mu X, Zhou H. Role of Interleukin 1 Receptor 2 in Kidney Disease. J Interferon Cytokine Res 2024; 44:170-177. [PMID: 38527174 DOI: 10.1089/jir.2023.0172] [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] [Indexed: 03/27/2024] Open
Abstract
The interleukin 1 (IL-1) family plays a significant role in the innate immune response. IL-1 receptor 2 (IL-1R2) is the decoy receptor of IL-1. It is a negative regulator that can be subdivided into membrane-bound and soluble types. IL-1R2 plays a role in the IL-1 family mainly through the following mechanisms: formation of inactive signaling complexes upon binding to the receptor auxiliary protein and inhibition of ligand IL-1 maturation. This review covers the roles of IL-1R2 in kidney disorders. Chronic kidney disease, acute kidney injury, lupus nephritis, IgA nephropathy, renal clear cell carcinoma, rhabdoid tumor of kidney, kidney transplantation, and kidney infection were all shown to have abnormal IL-1R2 expression. IL-1R2 may be a potential marker and a promising therapeutic target for kidney disease.
Collapse
Affiliation(s)
- Huiyue Hu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Aihua Wu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaodie Mu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hua Zhou
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
9
|
Steegh FMEG, Keijbeck AA, de Hoogt PA, Rademakers T, Houben AJHM, Reesink KD, Stehouwer CDA, Daemen MJAP, Peutz-Kootstra CJ. Capillary rarefaction: a missing link in renal and cardiovascular disease? Angiogenesis 2024; 27:23-35. [PMID: 37326760 DOI: 10.1007/s10456-023-09883-8] [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: 03/20/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023]
Abstract
Patients with chronic kidney disease (CKD) have an increased risk for cardiovascular morbidity and mortality. Capillary rarefaction may be both one of the causes as well as a consequence of CKD and cardiovascular disease. We reviewed the published literature on human biopsy studies and conclude that renal capillary rarefaction occurs independently of the cause of renal function decline. Moreover, glomerular hypertrophy may be an early sign of generalized endothelial dysfunction, while peritubular capillary loss occurs in advanced renal disease. Recent studies with non-invasive measurements show that capillary rarefaction is detected systemically (e.g., in the skin) in individuals with albuminuria, as sign of early CKD and/or generalized endothelial dysfunction. Decreased capillary density is found in omental fat, muscle and heart biopsies of patients with advanced CKD as well as in skin, fat, muscle, brain and heart biopsies of individuals with cardiovascular risk factors. No biopsy studies have yet been performed on capillary rarefaction in individuals with early CKD. At present it is unknown whether individuals with CKD and cardiovascular disease merely share the same risk factors for capillary rarefaction, or whether there is a causal relationship between rarefaction in renal and systemic capillaries. Further studies on renal and systemic capillary rarefaction, including their temporal relationship and underlying mechanisms are needed. This review stresses the importance of preserving and maintaining capillary integrity and homeostasis in the prevention and management of renal and cardiovascular disease.
Collapse
Affiliation(s)
- Floor M E G Steegh
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Anke A Keijbeck
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Patrick A de Hoogt
- Surgery, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Timo Rademakers
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Alfons J H M Houben
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Koen D Reesink
- Biomedical Engineering, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Mat J A P Daemen
- Department of Pathology, UMC Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Carine J Peutz-Kootstra
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
- Department of Pathology, Gelre Ziekenhuizen, Apeldoorn, The Netherlands.
- , Porthoslaan 39, 6213 CN, Maastricht, The Netherlands.
| |
Collapse
|
10
|
Schwarz N, Yadegari H. Potentials of Endothelial Colony-Forming Cells: Applications in Hemostasis and Thrombosis Disorders, from Unveiling Disease Pathophysiology to Cell Therapy. Hamostaseologie 2023; 43:325-337. [PMID: 37857295 DOI: 10.1055/a-2101-5936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Endothelial colony-forming cells (ECFCs) are endothelial progenitor cells circulating in a limited number in peripheral blood. They can give rise to mature endothelial cells (ECs) and, with intrinsically high proliferative potency, contribute to forming new blood vessels and restoring the damaged endothelium in vivo. ECFCs can be isolated from peripheral blood or umbilical cord and cultured to generate large amounts of autologous ECs in vitro. Upon differentiation in culture, ECFCs are excellent surrogates for mature ECs showing the same phenotypic, genotypic, and functional features. In the last two decades, the ECFCs from various vascular disease patients have been widely used to study the diseases' pathophysiology ex vivo and develop cell-based therapeutic approaches, including vascular regenerative therapy, tissue engineering, and gene therapy. In the current review, we will provide an updated overview of past studies, which have used ECFCs to elucidate the molecular mechanisms underlying the pathogenesis of hemostatic disorders in basic research. Additionally, we summarize preceding studies demonstrating the utility of ECFCs as cellular tools for diagnostic or therapeutic clinical applications in thrombosis and hemostasis.
Collapse
Affiliation(s)
- Nadine Schwarz
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Hamideh Yadegari
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
11
|
Spinelli FR, Berti R, Farina G, Ceccarelli F, Conti F, Crescioli C. Exercise-induced modulation of Interferon-signature: a therapeutic route toward management of Systemic Lupus Erythematosus. Autoimmun Rev 2023; 22:103412. [PMID: 37597604 DOI: 10.1016/j.autrev.2023.103412] [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: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Systemic Lupus Erythematosus (SLE) is a multisystemic autoimmune disorder characterized by flares-ups/remissions with a complex clinical picture related to disease severity and organ/tissue injury, which, if left untreated, may result in permanent damage. Enhanced fatigue and pain perception, worsened quality of life (QoL) and outcome are constant, albeit symptoms may differ. An aberrant SLE immunoprofiling, note as "interferon (IFN)α-signature", is acknowledged to break immunotolerance. Recently, a deregulated "IFNγ-signature" is suggested to silently precede/trigger IFNα profile before clinical manifestations. IFNα- and IFNγ-over-signaling merge in cytokine/chemokine overexpression exacerbating autoimmunity. Remission achievement and QoL improvement are the main goals. The current therapy (i.e., corticosteroids, immunosuppressants) aims to downregulate immune over-response. Exercise could be a safe treatment due to its ever-emerging ability to shape and re-balance immune system without harmful side-effects; in addition, it improves cardiorespiratory capacity and musculoskeletal strength/power, usually impaired in SLE. Nevertheless, exercise is not yet included in SLE care plans. Furthermore, due to the fear to worsening pain/fatigue, SLE subjects experience kinesiophobia and sedentary lifestyle, worsening physical health. Training SLE patients to exercise is mandatory to fight inactive behavior and ameliorate health. This review aims to focus the attention on the role of exercise as a non-pharmacological therapy in SLE, considering its ability to mitigate IFN-signature and rebalance (auto)immune response. To this purpose, the significance of IFNα- and IFNγ-signaling in SLE etiopathogenesis will be addressed first and discussed thereafter as biotarget of exercise. Comments are addressed on the need to make aware all SLE care professional figures to promote exercise for health patients.
Collapse
Affiliation(s)
- Francesca Romana Spinelli
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Riccardo Berti
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy
| | - Gabriele Farina
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy
| | - Fulvia Ceccarelli
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Fabrizio Conti
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Clara Crescioli
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy.
| |
Collapse
|
12
|
Cui G, Liu H, Laugsand JB. Endothelial cells-directed angiogenesis in colorectal cancer: Interleukin as the mediator and pharmacological target. Int Immunopharmacol 2023; 114:109525. [PMID: 36508917 DOI: 10.1016/j.intimp.2022.109525] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 12/14/2022]
Abstract
Enhanced angiogenesis is a cancer hallmark and critical for colorectal cancer (CRC) invasion and metastasis. Upon exposure to proangiogenic factors, therefore, targeting tumor-associated proangiogenic factors/receptors hold great promise as a therapeutic modality to treat CRC, particularly metastatic CRC. Accumulating evidence from numerous studies suggests that tumor endothelial cells (ECs) are not only the target of proangiogenic factors, but also function as the cellular source of proangiogenic factors. Studies showed that ECs can produce different proangiogenic factors to participate in the regulation of angiogenesis process, in which ECs-derived interleukins (ILs) show a potential stimulatory effect on angiogenesis via either an direct action on their receptors expressed on progenitor of ECs or an indirect way through enhanced production of other proangiogenic factors. Although a great deal of attention is given to the effects of tumor-derived and immune cell-derived ILs, few studies describe the potential effects of vascular ECs-derived ILs on the tumor angiogenesis process. This review provides an updated summary of available information on proangiogenic ILs, such as IL-1, IL-6, IL-8, IL-17, IL-22, IL-33, IL-34, and IL-37, released by microvascular ECs as potential drivers of the tumor angiogenesis process and discusses their potential as a novel candidate for antiangiogenic target for the treatment of CRC patients.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Faculty of Health Science, Nord University, Campus Levanger, Norway.
| | - Hanzhe Liu
- School of Stomatology, Wuhan University, Wuhan, China.
| | | |
Collapse
|
13
|
Guzmán-Martínez G, Marañón C. Immune mechanisms associated with cardiovascular disease in systemic lupus erythematosus: A path to potential biomarkers. Front Immunol 2022; 13:974826. [PMID: 36420265 PMCID: PMC9677819 DOI: 10.3389/fimmu.2022.974826] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/13/2022] [Indexed: 10/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) patients display an increased risk of cardiovascular disease (CVD). With the improved clinical management of other classical severe manifestation of the disease, CVD is becoming one of the most relevant complications of SLE, and it is an important factor causing morbidity and mortality. Several immune constituents have been shown to be involved in the pathogenesis of atherosclerosis and endothelial damage in SLE patients, including specific circulating cell populations, autoantibodies, and inflammatory mediators. In this review, we summarize the presentation of CVD in SLE and the role of the autoimmune responses present in SLE patients in the induction of atherogenesis, endothelial impairment and cardiac disease. Additionally, we discuss the utility of these immune mediators as early CVD biomarkers and targets for clinical intervention in SLE patients.
Collapse
Affiliation(s)
- Gabriela Guzmán-Martínez
- Atrys Health, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
- Department of Cardiology, La Paz University Hospital, IdiPaz, Madrid, Spain
| | - Concepción Marañón
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| |
Collapse
|
14
|
Lemmer D, Schmidt J, Kummer K, Lemmer B, Wrede A, Seitz C, Balcarek P, Schwarze K, Müller GA, Patschan D, Patschan S. Impairment of muscular endothelial cell regeneration in dermatomyositis. Front Neurol 2022; 13:952699. [PMID: 36330424 PMCID: PMC9623165 DOI: 10.3389/fneur.2022.952699] [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: 05/25/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
Background and aim Inflammatory myopathies are heterogeneous in terms of etiology, (immuno)pathology, and clinical findings. Endothelial cell injury, as it occurs in DM, is a common feature of numerous inflammatory and non-inflammatory vascular diseases. Vascular regeneration is mediated by both local and blood-derived mechanisms, such as the mobilization and activation of so-called proangiogenic cells (PACs) or early endothelial progenitor cells (eEPCs). The current study aimed to evaluate parameters of eEPC integrity in dermatomyositis (DM), compared to necrotizing myopathy (NM) and to non-myopathic controls. Methods Blood samples from DM and NM patients were compared to non-myositis controls and analyzed for the following parameters: circulating CD133+/VEGFR-2+ cells, number of colony-forming unit endothelial cells (CFU-ECs), concentrations of angiopoietin 1, vascular endothelial growth factor (VEGF), and CXCL-16. Muscle biopsies from DM and NM subjects underwent immunofluorescence analysis for CXCR6, nestin, and CD31 (PECAM-1). Finally, myotubes, derived from healthy donors, were stimulated with serum samples from DM and NM patients, subsequently followed by RT-PCR for the following candidates: IL-1β, IL-6, nestin, and CD31. Results Seventeen (17) DM patients, 7 NM patients, and 40 non-myositis controls were included. CD133+/VEGFR-2+ cells did not differ between the groups. Both DM and NM patients showed lower CFU-ECs than controls. In DM, intramuscular CD31 abundances were significantly reduced, which indicated vascular rarefaction. Muscular CXCR6 was elevated in both diseases. Circulating CXCL-16 was higher in DM and NM in contrast, compared to controls. Serum from patients with DM but not NM induced a profound upregulation of mRNS expression of CD31 and IL-6 in cultured myotubes. Conclusion Our study demonstrates the loss of intramuscular microvessels in DM, accompanied by endothelial activation in DM and NM. Vascular regeneration was impaired in DM and NM. The findings suggest a role for inflammation-associated vascular damage in the pathogenesis of DM.
Collapse
Affiliation(s)
- D. Lemmer
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
- Immanuel Krankenhaus Berlin, Medical Center of Rheumatology Berlin-Buch, Berlin, Germany
| | - J. Schmidt
- Department of Neurology and Pain Treatment, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - K. Kummer
- Department of Neurology, Neuromuscular Center, University Medical Center Göttingen, Göttingen, Germany
| | - B. Lemmer
- Department of Physics, Georg-August-University Göttingen, Göttingen, Germany
| | - A. Wrede
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - C. Seitz
- Department of Dermatology, Allergology and Venereology, University Medical Center Göttingen, Göttingen, Germany
| | - P. Balcarek
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Göttingen, Göttingen, Germany
- Arcus Klinik, Pforzheim, Germany
| | - K. Schwarze
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - G. A. Müller
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - D. Patschan
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Department of Medicine 1, Cardiology, Angiology, and Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Branderburg, Germany
| | - S. Patschan
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Department of Medicine 1, Cardiology, Angiology, and Nephrology, University Hospital Brandenburg of the Brandenburg Medical School Theodor Fontane, Branderburg, Germany
- *Correspondence: S. Patschan
| |
Collapse
|
15
|
Zhou X, Zhang Y, Wang N. Systematic identification of key extracellular proteins as the potential biomarkers in lupus nephritis. Front Immunol 2022; 13:915784. [PMID: 35967373 PMCID: PMC9366080 DOI: 10.3389/fimmu.2022.915784] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/01/2022] [Indexed: 11/21/2022] Open
Abstract
Background Lupus nephritis (LN) is the most common and severe clinical manifestation of systemic lupus erythematosus (SLE) with considerable morbidity/mortality and limited treatment options. Since kidney biopsy is a relative hysteretic indicator, it is indispensable to investigate potential biomarkers for early diagnosis and predicting clinical outcomes of LN patients. Extracellular proteins may become the promising biomarkers by the secretion into body fluid. Our study linked extracellular proteins with lupus nephritis to identify the emerging biomarkers. Methods The expression profiling data were acquired from the Gene Expression Omnibus (GEO) database. Meanwhile, the two gene lists encoding extracellular proteins were collected from the Human Protein Atlas (HPA) and UniProt database. Subsequently, the extracellular protein-differentially expressed genes (EP-DEGs) were screened out, and the key EP-DEGs were determined by MCODE, MCC, and Degree methods via the protein–protein interaction (PPI) network. The expression level, immune characteristics, and diagnostic value of these candidate biomarkers were investigated. Finally, the Nephroseq V5 tool was applied to evaluate the clinical significance of the key EP-DEGs. Results A total of 164 DEGs were acquired by comparing LN samples with healthy controls based on GSE32591 datasets. Then, 38 EP-DEGs were screened out through the intersection between DEGs and extracellular protein gene lists. Function enrichment analysis indicated that these EP-DEGs might participate in immune response and constitute the extracellular matrix. Four key EP-DEGs (LUM, TGFBI, COL1A2, and POSTN) were eventually identified as candidate biomarkers, and they were all overexpressed in LN samples. Except that LUM expression was negatively correlated with most of the immune regulatory genes, there was a positive correlation between the remaining three biomarkers and the immune regulatory genes. In addition, these biomarkers had high diagnostic value, especially the AUC value of the LUM–TGFBI combination which reached almost 1 (AUC = 0.973), demonstrating high accuracy in distinguishing LN from controls. Finally, we found a meaningful correlation of these biomarkers with sex, WHO class, and renal function such as glomerular filtration rate (GFR), serum creatinine level, and proteinuria. Conclusion In summary, our study comprehensively identified four key EP-DEGs exerting a vital role in LN diagnosis and pathogenesis and serving as promising therapeutic targets.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, China
- Haihe Hospital, Tianjin University, Tianjin, China
- Haihe Clinical School, Tianjin Medical University, Tianjin, China
| | - Yuefeng Zhang
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, China
- Haihe Hospital, Tianjin University, Tianjin, China
- Haihe Clinical School, Tianjin Medical University, Tianjin, China
| | - Ning Wang
- Medical Department, The Third Central Hospital of Tianjin, Tianjin, China
- *Correspondence: Ning Wang,
| |
Collapse
|
16
|
Alduraibi F, Fatima H, Hamilton JA, Chatham WW, Hsu HC, Mountz JD. Lupus nephritis correlates with B cell interferon-β, anti-Smith, and anti-DNA: a retrospective study. Arthritis Res Ther 2022; 24:87. [PMID: 35436902 PMCID: PMC9014622 DOI: 10.1186/s13075-022-02766-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/18/2022] [Indexed: 12/04/2022] Open
Abstract
Background In systemic lupus erythematosus (SLE), detection of interferon-β (IFNβ) in B cells was found to be most prominent in patients with high anti-Smith (Sm) and renal disease, but a mechanistic connection was not clear. The objective of the present study is to determine the association of IFNβ in peripheral blood naïve B cells with the histopathological features of lupus nephritis (LN). Methods The percentage of IFNβ+ cells in IgD+CD27− naïve CD19+ B cells (B cell IFNβ) among peripheral blood mononuclear cells (PBMCs) from 80 SLE patients were analyzed using flow cytometry. Serological and clinical data were collected. The correlations of B cell IFNβ with LN classification and with histopathological findings (light, electron, and immunofluorescence [IF] microscopic analyses for deposition of IgM, IgG, IgA, C1q, and C3) were determined in 23 available biopsy specimens. Results B cell IFNβ is positively associated with anti-Sm (p = 0.001), anti-DNA (p = 0.013), and LN (p < 0.001) but was negatively associated with oral/nasal ulcer (p = 0.003) and photosensitivity (p = 0.045). B cell IFNβ positively correlated with immune complex (IC) deposit in the glomerular basement membrane (GBM) (p = 0.002) but not in the mesangial (p = 0.107) or tubular region (p = 0.313). Patients with high B cell IFNβ had statistically increased development of the proliferative LN (Classes III, IV and/or V), compared to patients with low B cell IFNβ (p < 0.0001). Histopathological features positively associated with increased B cell IFNβ included active glomerular lesions as determined by fibrocellular crescents (p = 0.023), chronic glomerular lesions indicated by segmental sclerosis (p = 0.033), and a membranous pattern of renal damage indicated by spike/holes (p = 0.015). Conclusion B cell IFNβ correlates with history of severe LN, glomerular basement membrane (GBM) IC deposition, and anatomical features of both active and chronic glomerular lesions.
Collapse
Affiliation(s)
- Fatima Alduraibi
- Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA.,Medicine Service, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.,Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Huma Fatima
- Division of Anatomic Pathology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennie A Hamilton
- Department of Medicine, University of Tennessee Health Science Center, 920 Madison Ave, Memphis, TN, 38163, USA
| | - W Winn Chatham
- Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hui-Chen Hsu
- Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - John D Mountz
- Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA. .,Medicine Service, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
17
|
Mak A, Chan JKY. Endothelial function and endothelial progenitor cells in systemic lupus erythematosus. Nat Rev Rheumatol 2022; 18:286-300. [PMID: 35393604 DOI: 10.1038/s41584-022-00770-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
Abstract
The observations that traditional cardiovascular disease (CVD) risk factors fail to fully account for the excessive cardiovascular mortality in patients with systemic lupus erythematosus (SLE) compared with the general population have prompted in-depth investigations of non-traditional, SLE-related risk factors that contribute to cardiovascular complications in patients with SLE. Of the various perturbations of vascular physiology, endothelial dysfunction, which is believed to occur in the earliest step of atherosclerosis, has been extensively investigated for its contribution to CVD risk in SLE. Endothelial progenitor cells (EPCs), which play a crucial part in vascular repair, neovascularization and maintenance of endothelial function, are quantitatively and functionally reduced in patients with SLE. Yet, the lack of a unified definition of EPCs, standardization of the quantity and functional assessment of EPCs as well as endothelial function measurement pose challenges to the translation of endothelial function measurements and EPC levels into prognostic markers for CVD in patients with SLE. This Review discusses factors that contribute to CVD in SLE, with particular focus on how endothelial function and EPCs are evaluated currently, and how EPCs are quantitatively and functionally altered in patients with SLE. Potential strategies for the use of endothelial function measurements and EPC quantification as prognostic markers of CVD in patients with SLE, and the limitations of their prognostication potential, are also discussed.
Collapse
Affiliation(s)
- Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore, Singapore.
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Academic Clinical Programme in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
18
|
Lodi L, Mastrolia MV, Bello F, Rossi GM, Angelotti ML, Crow YJ, Romagnani P, Vaglio A. Type I interferon-related kidney disorders. Kidney Int 2022; 101:1142-1159. [PMID: 35339535 DOI: 10.1016/j.kint.2022.02.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/04/2022] [Accepted: 02/22/2022] [Indexed: 12/15/2022]
Abstract
Type I interferon (IFN-I) mediates tissue damage in a wide range of kidney disorders, directly affecting the biology and function of several renal cell types including podocytes, mesangial, endothelial and parietal epithelial cells (PECs).Enhanced IFN-I signalling is observed in the context of viral infections, autoimmunity (e.g., systemic lupus erythematosus, SLE), and the type 1 interferonopathies (T1Is), rare monogenic disorders characterised by constitutive activation of the IFN-I pathway. All of these IFN I-related disorders can cause renal dysfunction, and share pathogenic and histopathological features. Collapsing glomerulopathy, a histopathological lesion characterised by podocyte loss, collapse of the vascular tuft and PEC proliferation, is commonly associated with viral infections, has been described in T1Is such as Aicardi-Goutières syndrome and STING-associated vasculopathy with onset in infancy (SAVI), and can also be induced by recombinant IFN-therapy. In all of these conditions, podocytes and PECs seem to be the primary target of IFN I-mediated damage. Additionally, immune-mediated glomerular injury is common to viral infections, SLE, and T1Is such as COPA syndrome and DNASE1L3 deficiency, diseases in which IFN-I apparently promotes immune-mediated kidney injury. Finally, kidney pathology primarily characterised by vascular lesions (e.g., thrombotic microangiopathy, vasculitis) is a hallmark of the T1I ADA2 deficiency as well as of SLE, viral infections and IFN-therapy.Defining the nosology, pathogenic mechanisms and histopathological patterns of IFN I-related kidney disorders has diagnostic and therapeutic implications, especially considering the likely near-term availability of novel drugs targeting the IFN-I pathway.
Collapse
Affiliation(s)
- Lorenzo Lodi
- Department of Health Sciences, University of Firenze; Immunology Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy
| | - Maria V Mastrolia
- Rheumatology Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy
| | - Federica Bello
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | | | - Maria L Angelotti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK; Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Université de Paris, Paris, France
| | - Paola Romagnani
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy; Nephrology and Dialysis Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy
| | - Augusto Vaglio
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy; Nephrology and Dialysis Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy.
| |
Collapse
|
19
|
Oates JC, Russell DL, Van Beusecum JP. Endothelial cells: potential novel regulators of renal inflammation. Am J Physiol Renal Physiol 2022; 322:F309-F321. [PMID: 35129369 PMCID: PMC8897017 DOI: 10.1152/ajprenal.00371.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Substantial evidence has supported the role of endothelial cell (EC) activation and dysfunction in the development of hypertension, chronic kidney disease (CKD), and lupus nephritis (LN). In both humans and experimental models of hypertension, CKD, and LN, ECs become activated and release potent mediators of inflammation including cytokines, chemokines, and reactive oxygen species that cause EC dysfunction, tissue damage, and fibrosis. Factors that activate the endothelium include inflammatory cytokines, mechanical stretch, and pathological shear stress. These signals can activate the endothelium to promote upregulation of adhesion molecules, such as intercellular adhesion molecule-1 and vascular cell adhesion molecule-1, which promote leukocyte adhesion and migration to the activated endothelium. More importantly, it is now recognized that some of these signals may in turn promote endothelial antigen presentation through major histocompatibility complex II. In this review, we will consider in-depth mechanisms of endothelial activation and the novel mechanism of endothelial antigen presentation. Moreover, we will discuss these proinflammatory events in renal pathologies and consider possible new therapeutic approaches to limit the untoward effects of endothelial inflammation in hypertension, CKD, and LN.
Collapse
Affiliation(s)
- Jim C. Oates
- 1Ralph H. Johnson Veteran Affairs Medical Center, Charleston, South Carolina,2Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dayvia L. Russell
- 2Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Justin P. Van Beusecum
- 1Ralph H. Johnson Veteran Affairs Medical Center, Charleston, South Carolina,3Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
20
|
Nettersheim FS, Picard FSR, Hoyer FF, Winkels H. Immunotherapeutic Strategies in Cancer and Atherosclerosis-Two Sides of the Same Coin. Front Cardiovasc Med 2022; 8:812702. [PMID: 35097027 PMCID: PMC8792753 DOI: 10.3389/fcvm.2021.812702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
The development and clinical approval of immunotherapies has revolutionized cancer therapy. Although the role of adaptive immunity in atherogenesis is now well-established and several immunomodulatory strategies have proven beneficial in preclinical studies, anti-atherosclerotic immunotherapies available for clinical application are not available. Considering that adaptive immune responses are critically involved in both carcinogenesis and atherogenesis, immunotherapeutic approaches for the treatment of cancer and atherosclerosis may exert undesirable but also desirable side effects on the other condition, respectively. For example, the high antineoplastic efficacy of immune checkpoint inhibitors, which enhance effector immune responses against tumor cells by blocking co-inhibitory molecules, was recently shown to be constrained by substantial proatherogenic properties. In this review, we outline the specific role of immune responses in the development of cancer and atherosclerosis. Furthermore, we delineate how current cancer immunotherapies affect atherogenesis and discuss whether anti-atherosclerotic immunotherapies may similarly have an impact on carcinogenesis.
Collapse
Affiliation(s)
- Felix Sebastian Nettersheim
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Friedrich Felix Hoyer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
21
|
Blanco LP, Wang X, Carlucci PM, Torres-Ruiz JJ, Romo-Tena J, Sun HW, Hafner M, Kaplan MJ. RNA Externalized by Neutrophil Extracellular Traps Promotes Inflammatory Pathways in Endothelial Cells. Arthritis Rheumatol 2021; 73:2282-2292. [PMID: 33983685 PMCID: PMC8589882 DOI: 10.1002/art.41796] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 04/29/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) are extracellular lattices composed of nucleic material bound to neutrophil granule proteins. NETs may play pathogenic roles in the development and severity of autoimmune diseases such as systemic lupus erythematosus (SLE), at least in part, through induction of type I interferon (IFN) responses via externalization of oxidized immunostimulatory DNA. A distinct subset of SLE proinflammatory neutrophils (low-density granulocytes [LDGs]) displays enhanced ability to form proinflammatory NETs that damage the vasculature. We undertook this study to assess whether NET-bound RNA can contribute to inflammatory responses in endothelial cells (ECs) and the pathways that mediate this effect. METHODS Expression of newly synthesized and total RNA was quantified in NETs from healthy controls and lupus patients. The ability of ECs to take up NET-bound RNA and downstream induction of type I IFN responses were quantified. RNAs present in NETs were sequenced and specific small RNAs were tested for induction of endothelial type I IFN pathways. RESULTS NETs extruded RNA that was internalized by ECs, and this was enhanced when NET-bound nucleic acids were oxidized, particularly in lupus LDG-derived NETs. Internalization of NET-bound RNA by ECs was dependent on endosomal Toll-like receptors (TLRs) and the actin cytoskeleton and induced type I IFN-stimulated genes (ISGs). This ISG induction was dependent on NET-associated microRNA let-7b, a small RNA expressed at higher levels in LDG-derived NETs, which acted as a TLR-7 agonist. CONCLUSION These findings highlight underappreciated roles for small RNAs externalized in NETs in the induction of proinflammatory responses in vascular cells, with implications for lupus vasculopathy.
Collapse
Affiliation(s)
- Luz P. Blanco
- Systemic Autoimmunity Branch, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Xinghao Wang
- Systemic Autoimmunity Branch, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Philip M. Carlucci
- Systemic Autoimmunity Branch, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jose Jiram Torres-Ruiz
- Systemic Autoimmunity Branch, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jorge Romo-Tena
- Systemic Autoimmunity Branch, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Medical Science PhD Program, School of Medicine, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Markus Hafner
- RNA Molecular Biology Group, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
22
|
The severity of glomerular endothelial cell injury is associated with infiltrating macrophage heterogeneity in endocapillary proliferative glomerulonephritis. Sci Rep 2021; 11:13339. [PMID: 34172770 PMCID: PMC8233400 DOI: 10.1038/s41598-021-92655-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/14/2021] [Indexed: 12/31/2022] Open
Abstract
Endocapillary proliferation occurs in various types of glomerulonephritis (GN), with varying prognoses. We examined 42 renal biopsy samples representing endocapillary proliferative lesions from post-streptococcal acute GN (PSAGN), Henoch–Schönlein purpura nephritis (HSPN), and lupus nephritis (LN). In PSAGN, the glomerular capillary network was maintained, although severe lesions displayed dots or short, curved lines, indicating CD34-positive capillaries and suggesting capillary obstruction. Conversely, patients with LN and HSPN displayed obstruction of CD34-positive capillaries with dissociation from the glomerular basement membrane even in mild lesions. According to computer-assisted morphologic analysis, the cell density did not differ between the diseases. However, in PSAGN, the number of capillary loops was significantly increased, with a larger glomerular capillary luminal area than in the other groups. In addition, the number and frequency of CD163-positive cells (M2 macrophages) tended to be higher in PSAGN, while there were no significant differences in the number of CD68-positive (total) macrophages. These results indicate that in PSAGN, endothelial cell damage is less severe, and angiogenesis may be promoted. The severity of endothelial cell injury in each disease may be associated with differences in infiltrating inflammatory cell phenotypes.
Collapse
|
23
|
Ding X, Ren Y, He X. IFN-I Mediates Lupus Nephritis From the Beginning to Renal Fibrosis. Front Immunol 2021; 12:676082. [PMID: 33959133 PMCID: PMC8093624 DOI: 10.3389/fimmu.2021.676082] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Lupus nephritis (LN) is a common complication of systemic lupus erythematosus (SLE) and a major risk factor for morbidity and mortality. The abundant cell-free nucleic (DNA/RNA) in SLE patients, especially dsDNA, is a key substance in the pathogenesis of SLE and LN. The deposition of DNA/RNA-immune complexes (DNA/RNA-ICs) in the glomerulus causes a series of inflammatory reactions that lead to resident renal cell disturbance and eventually renal fibrosis. Cell-free DNA/RNA is the most effective inducer of type I interferons (IFN-I). Resident renal cells (rather than infiltrating immune cells) are the main source of IFN-I in the kidney. IFN-I in turn damages resident renal cells. Not only are resident renal cells victims, but also participants in this immunity war. However, the mechanism for generation of IFN-I in resident renal cells and the pathological mechanism of IFN-I promoting renal fibrosis have not been fully elucidated. This paper reviews the latest epidemiology of LN and its development process, discusses the mechanism for generation of IFN-I in resident renal cells and the role of IFN-I in the pathogenesis of LN, and may open a new perspective for the treatment of LN.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Ren
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Pediatric Internal Medicine Department, Haikou Maternal and Child Health Hospital, Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Type I Interferon as cardiovascular risk factor in systemic and cutaneous lupus erythematosus: A systematic review. Autoimmun Rev 2021; 20:102794. [PMID: 33722754 DOI: 10.1016/j.autrev.2021.102794] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Patients with systemic lupus erythematosus (SLE) have a high burden of cardiovascular disease (CVD) of multifactorial origin. The aim of this systematic review is to analyze the role of the interferon I (IFN-I) signature and fibroblast growth factor-23 (FGF-23) in patients with SLE or cutaneous lupus erythematosus (CLE) herein. MATERIALS AND METHODS We conducted a systematic literature search in PubMed and Scopus using keywords for major adverse cardiovascular events (MACE) and intermediate outcomes (endothelial dysfunction, subclinical atherosclerosis, platelet activation) associated with IFN-I or FGF-23 in patients with SLE and CLE. RESULTS 4745 citations were screened, of which 12 studies were included. IFN-I was associated with MACE in two third of the studies and the association was strongest for cardiac events. An association of IFN-I was found in all studies investigating impaired vascular function, but only in 50% (respectively 40%) of reports examining the relation of IFN-I and platelet activation (respectively subclinical atherosclerosis). Altogether the reports were of variable bias and quality due to high variability of examined IFN-I biomarkers and inconsistent results for different outcome measures. No studies investigating the cardiovascular risk of circulating IFN-I in CLE, nor FGF-23 in SLE or CLE were found. CONCLUSION Clinical studies measuring the association between IFN-I and direct / intermediate measures of CVD are rare and ambiguous in SLE and nonexistent in CLE, hampering a definite conclusion.
Collapse
|
25
|
van Nimwegen JF, van der Tuuk K, Liefers SC, Verstappen GM, Visser A, Wijnsma RF, Vissink A, Hollema H, Mourits MJE, Bootsma H, Kroese FGM. Vaginal dryness in primary Sjögren's syndrome: a histopathological case-control study. Rheumatology (Oxford) 2021; 59:2806-2815. [PMID: 32044981 PMCID: PMC7516088 DOI: 10.1093/rheumatology/keaa017] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/08/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE The aim was to study clinical, histopathological and immunological changes in the vagina and cervix of women with primary SS, which might explain vaginal dryness. METHODS We included 10 pre-menopausal female primary SS patients with vaginal dryness and 10 pre-menopausal controls undergoing a laparoscopic procedure. The vaginal health index was recorded. Multiplex immunoassays and flow cytometry were performed on endocervical swab and cervicovaginal lavage samples to evaluate cellular and soluble immune markers. Mid-vaginal and endocervical biopsies were taken and stained for various leucocyte markers, caldesmon (smooth muscle cells), avian V-ets erythroblastosis virus E26 oncogene homologue (ERG; endothelial cells) and anti-podoplanin (lymphatic endothelium). The number of positive pixels per square micrometre was calculated. RESULTS One patient was excluded because of Clamydia trachomatis, and two controls were excluded because of endometriosis observed during their laparoscopy. Vaginal health was impaired in primary SS. CD45+ cells were increased in vaginal biopsies of women with primary SS compared with controls. Infiltrates were predominantly located in the peri-epithelial region, and mostly consisted of CD3+ lymphocytes. In the endocervix, CD45+ infiltrates were present in patients and in controls, but a higher number of B lymphocytes was seen in primary SS. Vascular smooth muscle cells were decreased in the vagina of primary SS patients. No differences were found in leucocyte subsets in the vaginal and endocervical lumen. CXCL10 was increased in endocervical swab samples of primary SS patients. CONCLUSION Women with primary SS show impaired vaginal health and increased lymphocytic infiltration in the vagina compared with controls. Vaginal dryness in primary SS might be caused by vascular dysfunction, possibly induced by IFN-mediated pathways.
Collapse
Affiliation(s)
- Jolien F van Nimwegen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karin van der Tuuk
- Department of Obstetrics and Gynaecology, University of GroningenUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Silvia C Liefers
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gwenny M Verstappen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annie Visser
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robin F Wijnsma
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harry Hollema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marian J E Mourits
- Department of Obstetrics and Gynaecology, University of GroningenUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
26
|
Panopoulos S, Thomas K, Georgiopoulos G, Boumpas D, Katsiari C, Bertsias G, Drosos AA, Boki K, Dimitroulas T, Garyfallos A, Papagoras C, Katsimbri P, Tziortziotis A, Adamichou C, Kaltsonoudis E, Argyriou E, Vosvotekas G, Sfikakis PP, Vassilopoulos D, Tektonidou MG. Comparable or higher prevalence of comorbidities in antiphospholipid syndrome vs rheumatoid arthritis: a multicenter, case-control study. Rheumatology (Oxford) 2021; 60:170-178. [PMID: 32596727 DOI: 10.1093/rheumatology/keaa321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Evidence on comorbidity prevalence in antiphospholipid syndrome (APS) and its difference from high comorbidity burden rheumatic diseases is limited. Herein, we compare multiple comorbidities between APS and RA. METHODS A total of 326 patients from the Greek APS registry [237 women, mean age 48.7 (13.4) years, 161 primary APS (PAPS), 165 SLE-APS] were age/sex matched (1:2 ratio) with 652 patients from a Greek multicentre RA cohort of 3115 patients. Prevalence of cardiovascular (CV) risk factors, stroke, coronary artery disease (CAD), osteoporosis, diabetes mellitus (DM), chronic obstructive pulmonary disease (COPD), depression and neoplasms were compared between APS and RA patients using multivariate regression analysis. RESULTS Ηyperlipidemia and obesity (ΒΜΙ ≥ 30 kg/m2) were comparable while hypertension, smoking, stroke and CAD were more prevalent in APS compared with RA patients. Osteoporosis and depression were more frequent in APS, while DM, COPD and neoplasms did not differ between the two groups. Comparison of APS subgroups to 1:2 matched RA patients revealed that smoking and stroke were more prevalent in both PAPS and SLE-APS vs RA. Hypertension, CAD and osteoporosis were more frequent only in SLE-APS vs RA, whereas DM was less prevalent in PAPS vs RA. Hyperlipidaemia was independently associated with CV events (combined stroke and CAD) in PAPS and SLE-APS, while CS duration was associated with osteoporosis in SLE-APS. CONCLUSION Comorbidity burden in APS (PAPS and SLE-APS) is comparable or higher than that in RA, entailing a high level of diligence for CV risk prevention, awareness for depression and CS exposure minimization.
Collapse
Affiliation(s)
- Stylianos Panopoulos
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| | - Konstantinos Thomas
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| | - Georgios Georgiopoulos
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| | - Dimitrios Boumpas
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| | | | | | | | | | | | | | | | - Pelagia Katsimbri
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| | | | | | | | | | | | - Petros P Sfikakis
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| | - Dimitrios Vassilopoulos
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| | - Maria G Tektonidou
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens
| |
Collapse
|
27
|
Ding X, Xiang W, He X. IFN-I Mediates Dysfunction of Endothelial Progenitor Cells in Atherosclerosis of Systemic Lupus Erythematosus. Front Immunol 2020; 11:581385. [PMID: 33262760 PMCID: PMC7686511 DOI: 10.3389/fimmu.2020.581385] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, NHC Key Laboratory of Control of Tropical diseases (Hainan Medical University), Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Komici K, Faris P, Negri S, Rosti V, García-Carrasco M, Mendoza-Pinto C, Berra-Romani R, Cervera R, Guerra G, Moccia F. Systemic lupus erythematosus, endothelial progenitor cells and intracellular Ca2+ signaling: A novel approach for an old disease. J Autoimmun 2020; 112:102486. [DOI: 10.1016/j.jaut.2020.102486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 02/07/2023]
|
29
|
Tsai TY, Lee TH, Wang HH, Yang TH, Chang IJ, Huang YC. Serum Homocysteine, Folate, and Vitamin B 12 Levels in Patients with Systemic Lupus Erythematosus: A Meta-Analysis and Meta-Regression. J Am Coll Nutr 2020; 40:443-453. [PMID: 32702250 DOI: 10.1080/07315724.2020.1788472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE) have elevated cardiovascular risk. Hyperhomocysteinemia may be one of the contributing factors to this phenomenon. This study therefore aimed to compare the serum homocysteine levels and the levels of folate and vitamin B12, cofactors for homocysteine metabolism, between individuals with and without SLE. METHODS A literature search was performed in PubMed, Embase, and the Cochrane library (from inception to March 31, 2019). Studies comparing serum homocysteine, folate or vitamin B12 levels between individuals with and without SLE were selected. Of the 1040 screened studies, 50 studies met the inclusion criteria. RESULTS A total of 50 studies involving 4396 patients with SLE were included. Patients with SLE had a significantly higher serum level of homocysteine (standardized mean difference [SMD], 1.134; 95% CI, 0.795-1.474) and lower level of vitamin B12 (SMD, -0.359; 95% CI, -0.638 to -0.080) than controls. The folate level didn't differ markedly between SLE patients and the control group (SMD, -0.276; 95% CI, -0.674-0.123). Subgroup analysis showed consistent results in adult SLE patients. A random effects meta-regression analysis revealed a significantly inverse correlation between the SMD of homocysteine levels and C3 levels (coefficient, -0.0356, 95% CI, -0.054 to -0.0171; P < .001) and C4 levels (coefficient, -0.0876, 95% CI, -0.1407 to -0.0345; P = .0012). CONCLUSIONS Serum homocysteine levels were higher and vitamin B12 levels were lower among individuals with SLE than those without SLE. Physicians are encouraged to monitor these parameters and offer timely interventions for patients with SLE.
Collapse
Affiliation(s)
- Tsung-Yu Tsai
- Department of Dermatology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tsai-Hsien Lee
- Department of Dermatology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsiao-Han Wang
- Department of Dermatology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ting-Hua Yang
- Department of Dermatology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - I-Jing Chang
- Department of Dermatology, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Huang
- Department of Dermatology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
30
|
Interaction of miR-181b and IFNA1 Polymorphisms on the Risk of Systemic Lupus Erythematosus. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4757065. [PMID: 32382553 PMCID: PMC7196983 DOI: 10.1155/2020/4757065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/16/2020] [Indexed: 12/23/2022]
Abstract
Introduction A previous work has discovered that chromosome 1q32 locus linked to the risk of systemic lupus erythematosus (SLE) and miR-181b located on the susceptibility site with downregulation inversely correlating to its target molecular interferon alpha 1 (IFNA1). The purpose of this study was to investigate the association of miR-181b and IFNA1 polymorphisms with IS risk. Methods The miR-181b rs322931, IFNA1 rs1332190, and rs10811543 were genotyped using a Multiplex SNaPshot assay. miR-181b expression levels in plasma of SLE patients and controls were analyzed using quantitative PCR. Results The rs322931 CT, CT/TT, and T allele exerted an increased trend of SLE risk (CT vs. CC: adjusted OR = 1.71, 95% CI 1.16-2.50, P = 0.01; CT/TT vs. CC: adjusted OR = 1.45, 95% CI 1.08-1.95, P = 0.01; T vs. C: adjusted OR = 1.38, 95% CI 1.07-1.79, P = 0.01). Combined genotypes of the rs322931 CT/TT+rs1332190 TT and the rs322931 CC+rs10811543 AG/AA also revealed an increased risk of SLE. Gene-gene interaction analysis showed that a three-locus model consisting of rs322931, rs1332190, and rs10811543 attributed an increased risk of SLE. Further genotype-phenotype analysis revealed that rs322931 CT/TT carriers displayed lower levels of miR-181b. Conclusions These findings indicate that the miR-181b rs322931 may be singly and jointly responsible for the etiology of SLE by altering miR-181b expression.
Collapse
|
31
|
Protecting the kidney in systemic lupus erythematosus: from diagnosis to therapy. Nat Rev Rheumatol 2020; 16:255-267. [PMID: 32203285 DOI: 10.1038/s41584-020-0401-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2020] [Indexed: 12/20/2022]
Abstract
Lupus nephritis (LN) is a common manifestation of systemic lupus erythematosus that can lead to irreversible renal impairment. Although the prognosis of LN has improved substantially over the past 50 years, outcomes have plateaued in the USA in the past 20 years as immunosuppressive therapies have failed to reverse disease in more than half of treated patients. This failure might reflect disease complexity and heterogeneity, as well as social and economic barriers to health-care access that can delay intervention until after damage has already occurred. LN progression is still poorly understood and involves multiple cell types and both immune and non-immune mechanisms. Single-cell analysis of intrinsic renal cells and infiltrating cells from patients with LN is a new approach that will help to define the pathways of renal injury at a cellular level. Although many new immune-modulating therapies are being tested in the clinic, the development of therapies to improve regeneration of the injured kidney and to prevent fibrosis requires a better understanding of the mechanisms of LN progression. This mechanistic understanding, together with the development of clinical measures to evaluate risk and detect early disease and better access to expert health-care providers, should improve outcomes for patients with LN.
Collapse
|
32
|
Mariotte A, De Cauwer A, Po C, Abou-Faycal C, Pichot A, Paul N, Aouadi I, Carapito R, Frisch B, Macquin C, Chatelus E, Sibilia J, Armspach JP, Bahram S, Georgel P. A mouse model of MSU-induced acute inflammation in vivo suggests imiquimod-dependent targeting of Il-1β as relevant therapy for gout patients. Am J Cancer Res 2020; 10:2158-2171. [PMID: 32104502 PMCID: PMC7019178 DOI: 10.7150/thno.40650] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: The role of Monosodium Urate (MSU) crystals in gout pathophysiology is well described, as is the major impact of IL-1β in the inflammatory reaction that constitutes the hallmark of the disease. However, despite the discovery of the NLRP3 inflammasome and its role as a Pattern Recognition Receptor linking the detection of a danger signal (MSU) to IL-1β secretion in vitro, the precise mechanisms leading to joint inflammation in gout patients are still poorly understood. Methods: Acute urate crystal inflammation was obtained by subcutaneous injections of MSU crystals in mice. Symptoms were followed by scoring, cytokine quantification by ELISA and western blot, gene expression by RT-qPCR and RNAseq; Magnetic Resonance Imaging was also used to assess inflammation. Results: We provide an extensive clinical, biological and molecular characterization of an acute uratic inflammation mouse model which accurately mimics human gout. We report the efficacy of topical imiquimod treatment and its impact on Interferon-dependent down modulation of Il-1β gene expression in this experimental model. Conclusion: Our work reveals several key features of MSU-dependent inflammation and identifies novel therapeutic opportunities for gout patients.
Collapse
|
33
|
Zhang W, Borcherding N, Kolb R. IL-1 Signaling in Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:1-23. [PMID: 32060884 DOI: 10.1007/978-3-030-38315-2_1] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Interleukin 1 (IL-1) has long been known for its pleiotropic effects on inflammation that plays a complex, and sometimes contrasting, role in different stages of cancer development. As a major proinflammatory cytokine, IL-1β is mainly expressed by innate immune cells. IL-1α, however, is expressed by various cell types under physiological and pathological conditions. IL-1R1 is the main receptor for both ligands and is expressed by various cell types, including innate and adaptive immune cell types, epithelial cells, endothelial cells, adipocytes, chondrocytes, fibroblasts, etc. IL-1 and IL-1R1 receptor interaction leads to a set of common signaling pathways, mainly the NF-kB and MAP kinase pathways, as a result of complex positive and negative regulations. The variety of cell types with IL-1R1 expression dictates the role of IL-1 signaling at different stages of cancer, which under certain circumstances leads to contrasting roles in tumor development. Recent availability of IL-1R1 conditional knockout mouse model has made it possible to dissect the role of IL-1/IL-1R1 signaling transduction in different cell types within the tumor microenvironment. This chapter will focus on the role of IL-1/IL-1R1 in different cell types within the tumor microenvironment and discuss the potential of targeting this pathway in cancer therapy.
Collapse
Affiliation(s)
- Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
| | | | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| |
Collapse
|
34
|
Abstract
Renal microvascular lesions, common in lupus nephritis (LN), are associated with long-term poor outcomes. There are mainly five pathological types of renal microvascular lesions in LN: (1) vascular immune complex deposits (ICD), (2) arteriosclerosis (AS), (3) thrombotic microangiopathy (TMA), (4) non-inflammatory necrotizing vasculopathy (NNV), and (5) true renal vasculitis (TRV). The pathogenesis of renal microvascular lesions in LN remains to be elucidated. The activation and dysfunction of endothelial cells, in addition to the contribution of immune system dysfunction, especially the immune complex-induced vascular inflammation and antiphospholipid antibody-associated thrombotic events, are key mechanisms in the development of vascular lesions in LN that need to be further investigated. Alteration of the microvascular environment produces an acute immunological response that recruits immune cells, such as T cells, monocytes, and macrophages, which induces platelet aggregation with microthrombus formation. There is also increased cytotoxicity caused by cytokines produced by immune cells in the kidney. Identifying the mechanism underlying the pathogenesis of renal microvascular lesions in LN might provide potential targets for the development of novel therapies.
Collapse
Affiliation(s)
- Ying Ding
- Department of Nephrology, Peking University International Hospital, Beijing, PR. China.,Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Beijing, PR. China.,Key laboratory of Renal Disease, Ministry of Health of China, Beijing, PR. 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, Beijing, PR. China.,Key laboratory of Renal Disease, Ministry of Health of China, Beijing, PR. China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR. China
| | - Zhen Qu
- Department of Nephrology, Peking University International Hospital, Beijing, PR. China
| | - Feng Yu
- Department of Nephrology, Peking University International Hospital, Beijing, PR. China.,Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Beijing, PR. China.,Key laboratory of Renal Disease, Ministry of Health of China, Beijing, PR. China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR. China
| |
Collapse
|
35
|
The Role of Neutrophils and Neutrophil Extracellular Traps in Vascular Damage in Systemic Lupus Erythematosus. J Clin Med 2019; 8:jcm8091325. [PMID: 31466329 PMCID: PMC6780421 DOI: 10.3390/jcm8091325] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/16/2019] [Accepted: 08/25/2019] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune syndrome of unknown etiology, characterized by multi-organ inflammation and clinical heterogeneity. SLE affects mostly women and is associated with a high risk of cardiovascular disease. As the therapeutic management of SLE improved, a pattern of early atherosclerotic disease became one of the hallmarks of late disease morbidity and mortality. Neutrophils emerged as important players in SLE pathogenesis and they are associated with increased risk of developing atherosclerotic disease and vascular damage. Enhanced neutrophil extracellular trap (NET) formation was linked to vasculopathy in both SLE and non-SLE subjects and may promote enhanced coronary plaque formation and lipoprotein dysregulation. Foundational work provided insight into the complex relationship between NETs and immune and tissue resident cells within the diseased artery. In this review, we highlight the mechanistic link between neutrophils, NETs, and atherosclerosis within the context of both SLE and non-SLE subjects. We aim to identify actionable pathways that will drive future research toward translational therapeutics, with the ultimate goal of preventing early morbidity and mortality in SLE.
Collapse
|
36
|
Rönnblom L, Leonard D. Interferon pathway in SLE: one key to unlocking the mystery of the disease. Lupus Sci Med 2019; 6:e000270. [PMID: 31497305 PMCID: PMC6703304 DOI: 10.1136/lupus-2018-000270] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/19/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022]
Abstract
SLE is characterised by an activation of the interferon (IFN) system, which leads to an increased expression of IFN-regulated genes. The reasons behind the IFN signature in SLE are (1) the existence of endogenous IFN inducers, (2) activation of several IFN-producing cell types, (3) production of many different IFNs, (4) a genetic setup promoting IFN production and (5) deficient negative feedback mechanisms. The consequences for the immune system is a continuous stimulation to an immune response, and for the patient a number of different organ manifestations leading to typical symptoms for SLE. In the current review, we will present the existing knowledge of the IFN system and pathway activation in SLE. We will also discuss how this information can contribute to our understanding of both the aetiopathogenesis and some organ manifestations of the disease. We will put forward some issues that are unresolved and should be clarified in order to make a proper stratification of patients with SLE, which seems important when selecting a therapy aiming to downregulate the IFN system.
Collapse
Affiliation(s)
- Lars Rönnblom
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Dag Leonard
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
37
|
Kishi T, Chipman J, Evereklian M, Nghiem K, Stetler-Stevenson M, Rick ME, Centola M, Miller FW, Rider LG. Endothelial Activation Markers as Disease Activity and Damage Measures in Juvenile Dermatomyositis. J Rheumatol 2019; 47:1011-1018. [PMID: 31371656 DOI: 10.3899/jrheum.181275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Circulating endothelial cells (CEC), von Willebrand factor (vWF) antigen, P-selectin, and thrombomodulin are released from damaged endothelium, while decreases in circulating endothelial progenitor cells (CEPC) have been associated with poor vascular outcomes. We examined these markers in the peripheral blood of patients with juvenile dermatomyositis (JDM) and their correlations with disease assessments. METHODS Peripheral blood endothelial cells and biomarkers were assessed in 20 patients with JDM and matched healthy controls. CEC and CEPC were measured by flow cytometry, while vWF antigen and activity, factor VIII, P-selectin, and thrombomodulin were measured in plate-based assays. Disease activity and damage, nailfold capillary density, and brachial artery flow dilation were assessed. Serum cytokines/chemokines were measured by Luminex. RESULTS CEC, vWF antigen, factor VIII, and thrombomodulin, but not vWF activity, CEPC, or P-selectin, were elevated in the peripheral blood of patients with JDM. CEC correlated with pulmonary activity (rs = 0.56). The vWF antigen correlated with Patient's/Parent's Global, cutaneous, and extramuscular activity (rs = 0.47-0.54). CEPC negatively correlated with muscle activity and physical function (rs = -0.52 to -0.53). CEPC correlated inversely with endocrine damage. The vWF antigen and activity correlated with interleukin 10 and interferon-gamma inducible protein-10 (rs = 0.64-0.82). CONCLUSION Markers of endothelial injury are increased in patients with JDM and correlate with extramuscular activity. CEPC correlate inversely with muscle activity, suggesting a functional disturbance in repair mechanisms.
Collapse
Affiliation(s)
- Takayuki Kishi
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Jonathan Chipman
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Melvina Evereklian
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Khanh Nghiem
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Maryalice Stetler-Stevenson
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Margaret E Rick
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Michael Centola
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Frederick W Miller
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA.,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH
| | - Lisa G Rider
- From the Environmental Autoimmunity Group, Clinical Research Branch, US National Institute of Environmental Health Sciences, National Institutes of Health (NIH); Coagulation Laboratory, NIH Clinical Center; Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH, Bethesda, Maryland; Oklahoma Medical Research Foundation; Haus Bioceuticals Inc., Oklahoma City, Oklahoma, USA. .,T. Kishi, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; J. Chipman, MS, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; M. Evereklian, MSN, CPNP-BC, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; K. Nghiem, MS, Coagulation Laboratory, NIH Clinical Center; M. Stetler-Stevenson, MD, Laboratories of Molecular Biology and Pathology, National Cancer Institute, NIH; M.E. Rick, MD, Coagulation Laboratory, NIH Clinical Center; M. Centola, PhD, Oklahoma Medical Research Foundation, and Haus Bioceuticals Inc.; F.W. Miller, MD, PhD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH; L.G. Rider, MD, Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH.
| |
Collapse
|
38
|
Association between Type I interferon and depletion and dysfunction of endothelial progenitor cells in C57BL/6 mice deficient in both apolipoprotein E and Fas ligand. Curr Res Transl Med 2018; 66:71-82. [DOI: 10.1016/j.retram.2018.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 10/31/2017] [Accepted: 02/07/2018] [Indexed: 11/22/2022]
|
39
|
Liu Y, Seto NL, Carmona-Rivera C, Kaplan MJ. Accelerated model of lupus autoimmunity and vasculopathy driven by toll-like receptor 7/9 imbalance. Lupus Sci Med 2018; 5:e000259. [PMID: 29765617 PMCID: PMC5950641 DOI: 10.1136/lupus-2018-000259] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 12/30/2022]
Abstract
Objectives Activation of endosomal toll-like receptor (TLR)7 or TLR9 has been proposed as a critical step for the initiation and development of SLE. Traditional spontaneous lupus models normally introduce multiple risk alleles, thereby adding additional confounding factors. In the induced lupus models, the role of TLR9 remains unclear. In the present study, we explored the role of an imbalance between TLR7 and TLR9 pathways in the pathogenesis of lupus and its associated vasculopathy using the imiquimod model in TLR9 KO/B6 background. Methods Wild type (WT) and Tlr9-/- mice were epicutaneously treated with imiquimod cream 5% on both ears three times per week for indicated times. At euthanasia, mice were analysed for organ involvement, endothelium-dependent vasorelaxation, serum autoantibodies, and innate and adaptive immune responses. Results Compared with the lupus-like phenotype that develops in imiquimod-treated WT mice, Tlr9-/- mice exposed to imiquimod have increased severity of autoimmunity features and inflammatory phenotype that develops at earlier stages. These abnormalities are characterised by enhanced TLR7 expression and immune activation, increased immune complex deposition, Th1 T cells and dendritic cell kidney infiltration and significant impairments in endothelial function. Modulation of TLR7 expression was observed in the Tlr9-/- mice. Conclusions These findings further underscore the protective role of TLR9 in TLR7-driven autoimmunity and also in the development of vasculopathy, further strengthening the importance of tightly manipulating TLRs in putative therapeutic strategies. This study provides a new model of accelerated lupus phenotype driven by danger-associated molecular patterns.
Collapse
Affiliation(s)
- Yudong Liu
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nickie L Seto
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
40
|
Carlucci PM, Purmalek MM, Dey AK, Temesgen-Oyelakin Y, Sakhardande S, Joshi AA, Lerman JB, Fike A, Davis M, Chung JH, Playford MP, Naqi M, Mistry P, Gutierrez-Cruz G, Dell'Orso S, Naz F, Salahuddin T, Natarajan B, Manna Z, Tsai WL, Gupta S, Grayson P, Teague H, Chen MY, Sun HW, Hasni S, Mehta NN, Kaplan MJ. Neutrophil subsets and their gene signature associate with vascular inflammation and coronary atherosclerosis in lupus. JCI Insight 2018; 3:99276. [PMID: 29669944 DOI: 10.1172/jci.insight.99276] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/09/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is associated with enhanced risk of atherosclerotic cardiovascular disease not explained by Framingham risk score (FRS). Immune dysregulation associated to a distinct subset of lupus proinflammatory neutrophils (low density granulocytes; LDGs) may play key roles in conferring enhanced CV risk. This study assessed if lupus LDGs are associated with in vivo vascular dysfunction and inflammation and coronary plaque. METHODS SLE subjects and healthy controls underwent multimodal phenotyping of vascular disease by quantifying vascular inflammation (18F-fluorodeoxyglucose-PET/CT [18F-FDG-PET/CT]), arterial dysfunction (EndoPAT and cardio-ankle vascular index), and coronary plaque burden (coronary CT angiography). LDGs were quantified by flow cytometry. Cholesterol efflux capacity was measured in high-density lipoprotein-exposed (HDL-exposed) radioactively labeled cell lines. Whole blood RNA sequencing was performed to assess associations between transcriptomic profiles and vascular phenotype. RESULTS Vascular inflammation, arterial stiffness, and noncalcified plaque burden (NCB) were increased in SLE compared with controls even after adjustment for traditional risk factors. In SLE, NCB directly associated with LDGs and associated negatively with cholesterol efflux capacity in fully adjusted models. A neutrophil gene signature reflective of the most upregulated genes in lupus LDGs associated with vascular inflammation and NCB. CONCLUSION Individuals with SLE demonstrate vascular inflammation, arterial dysfunction, and NCB, which may explain the higher reported risk for acute coronary syndromes. The association of LDGs and neutrophil genes with vascular disease supports the hypothesis that distinct neutrophil subsets contribute to vascular damage and unstable coronary plaque in SLE. Results also support previous observations that neutrophils may disrupt HDL function and thereby promote atherogenesis. TRIAL REGISTRATION Clinicaltrials.gov NCT00001372FUNDING. Intramural Research Program NIAMS/NIH (ZIA AR041199) and Lupus Research Institute.
Collapse
Affiliation(s)
- Philip M Carlucci
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Monica M Purmalek
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Amit K Dey
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | | | | | - Aditya A Joshi
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Joseph B Lerman
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Alice Fike
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Michael Davis
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Jonathan H Chung
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Martin P Playford
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Mohammad Naqi
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Pragnesh Mistry
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | | | - Stefania Dell'Orso
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Faiza Naz
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Taufiq Salahuddin
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Balaji Natarajan
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Zerai Manna
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Wanxia L Tsai
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Sarthak Gupta
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Peter Grayson
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Heather Teague
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Marcus Y Chen
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Hong-Wei Sun
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Sarfaraz Hasni
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| | - Nehal N Mehta
- National Heart, Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) and
| |
Collapse
|
41
|
Littlejohn E, Marder W, Lewis E, Francis S, Jackish J, McCune WJ, Somers EC. The ratio of erythrocyte sedimentation rate to C-reactive protein is useful in distinguishing infection from flare in systemic lupus erythematosus patients presenting with fever. Lupus 2018; 27:1123-1129. [PMID: 29546774 DOI: 10.1177/0961203318763732] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Both C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) can be elevated in systemic lupus erythematosus (SLE) flare and infection, and are therefore of limited utility for distinguishing between the two conditions in febrile SLE patients. Methods A medical records review of hospitalizations (1997-2006) of SLE patients in the Michigan Lupus Cohort was performed. Eligible hospitalizations were those in which patients presented with a temperature of >100.3°F or with subjective fevers as a presenting complaint at admission. Detailed demographic, clinical, and laboratory data were collected. Multivariable logistic regression was used to examine the associations between ESR and CRP and the outcome of flare vs infection, adjusted for confounders. Results Among 557 SLE patients screened, there were 53 eligible hospitalizations (28 flares and 25 infections). Each unit increase in the ratio of ESR:CRP was associated with a 17% increase in the odds of fever being attributable to SLE flare compared to infection (OR 1.17, 95% CI 1.04, 1.31; p = 0.009), when adjusted for white blood cell count, SLE duration, sex, race, and age. ESR and CRP were not individually associated with flare vs infection when modeled with their ratio. Conclusions The ratio of ESR:CRP may provide diagnostic value beyond individual ESR and CRP levels in distinguishing flare vs infection in SLE patients presenting with fever.
Collapse
Affiliation(s)
- E Littlejohn
- 1 Division of Rheumatology, Department of Internal Medicine, 1259 University of Michigan , Ann Arbor, MI, USA
| | - W Marder
- 1 Division of Rheumatology, Department of Internal Medicine, 1259 University of Michigan , Ann Arbor, MI, USA.,2 Department of Obstetrics & Gynecology, 1259 University of Michigan , Ann Arbor, MI, USA
| | - E Lewis
- 1 Division of Rheumatology, Department of Internal Medicine, 1259 University of Michigan , Ann Arbor, MI, USA
| | - S Francis
- 1 Division of Rheumatology, Department of Internal Medicine, 1259 University of Michigan , Ann Arbor, MI, USA.,3 Orthopedics & Sports Medicine P.C., New Windsor, NY, USA
| | - J Jackish
- 1 Division of Rheumatology, Department of Internal Medicine, 1259 University of Michigan , Ann Arbor, MI, USA.,4 25214 Park Nicollet/Health Partners Methodist Hospital , St. Louis Park, MN, USA
| | - W J McCune
- 1 Division of Rheumatology, Department of Internal Medicine, 1259 University of Michigan , Ann Arbor, MI, USA
| | - E C Somers
- 1 Division of Rheumatology, Department of Internal Medicine, 1259 University of Michigan , Ann Arbor, MI, USA.,2 Department of Obstetrics & Gynecology, 1259 University of Michigan , Ann Arbor, MI, USA.,5 Department of Environmental Health Sciences, 1259 University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
42
|
Jia H, Thelwell C, Dilger P, Bird C, Daniels S, Wadhwa M. Endothelial cell functions impaired by interferon in vitro: Insights into the molecular mechanism of thrombotic microangiopathy associated with interferon therapy. Thromb Res 2018; 163:105-116. [PMID: 29407621 DOI: 10.1016/j.thromres.2018.01.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/22/2017] [Accepted: 01/22/2018] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Interferon (IFN)-α and IFN-β approved for treatment of chronic hepatitis C viral infection and multiple sclerosis respectively have been linked to thrombotic microangiopathy (TMA) affecting renal function. Since the molecular mechanisms underlying this severe complication remain largely unclear, we aimed to investigate whether IFN affects directly in vitro endothelial cell functions associated with angiogenesis and blood haemostasis, as well as endothelial cell-derived vasodilators of nitric oxide (NO) and prostacyclin. METHODS Proliferation and survival of human umbilical vein endothelial cells (HUVECs) were measured by BrdU incorporation and alamarBlue assays. Angiogenesis was evaluated in co-cultures of HUVECs and human dermal fibroblasts. Fibrinolysis molecules were measured with ELISA. NO and prostacyclin were measured using a fluorescent NO-specific probe and a competitive enzyme immunoassay, respectively. RESULTS HUVEC proliferation was dose-dependently inhibited by IFN-β1a and IFN-β1b, but not by IFN-α2a and IFN-α2b. Consistently, IFN-β1a and IFN-β1b also reduced survival of HUVECs, but this again was not observed with IFN-α. However, both IFN subtypes inhibited VEGF-induced development of capillary-like structures, but the effect of IFN-α was less potent than IFN-β. In addition, both IFN subtypes upregulated interferon inducible protein 10 production from treated co-cultures while suppressing angiogenesis. Furthermore, intracellular NO generation was reduced by IFN-α2a and IFN-β1a, whereas prostacyclin release from HUVECs was not affected by IFN. Importantly, both IFN-β1a- and IFN-β1b-treated HUVECs showed a marked reduction in urokinase-type plasminogen activator release and a much greater secretion of plasminogen activator inhibitor-1 than tissue-type plasminogen activator compared with untreated cells, suggesting decreased fibrinolytic activity. IFN-α, however was less effective in modulating the fibrinolysis system. CONCLUSIONS We demonstrate the detrimental effects of IFN on endothelial cell functions mediated with angiogenesis and fibrinolysis, which could potentially cause the loss of physiological endothelium thromboresistance and facilitate the development of vascular complications in a clinical setting. Mechanistically, our findings have implications for understanding how IFN therapy can foster the development of TMA.
Collapse
Affiliation(s)
- Haiyan Jia
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom.
| | - Craig Thelwell
- Section of Haemostasis, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Paula Dilger
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Chris Bird
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Sarah Daniels
- Section of Haemostasis, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Meenu Wadhwa
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| |
Collapse
|
43
|
Tydén H, Lood C, Gullstrand B, Nielsen CT, Heegaard NHH, Kahn R, Jönsen A, Bengtsson AA. Endothelial dysfunction is associated with activation of the type I interferon system and platelets in patients with systemic lupus erythematosus. RMD Open 2017; 3:e000508. [PMID: 29119007 PMCID: PMC5663269 DOI: 10.1136/rmdopen-2017-000508] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/17/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022] Open
Abstract
Objectives Endothelial dysfunction may be connected to cardiovascular disease (CVD) in systemic lupus erythematosus (SLE). Type I interferons (IFNs) are central in SLE pathogenesis and are suggested to induce both endothelial dysfunction and platelet activation. In this study, we investigated the interplay between endothelial dysfunction, platelets and type I IFN in SLE. Methods We enrolled 148 patients with SLE and 79 sex-matched and age-matched healthy controls (HCs). Type I IFN activity was assessed with a reporter cell assay and platelet activation by flow cytometry. Endothelial dysfunction was assessed using surrogate markers of endothelial activation, soluble vascular cell adhesion molecule-1 (sVCAM-1) and endothelial microparticles (EMPs), and finger plethysmograph to determine Reactive Hyperaemia Index (RHI). Results In patients with SLE, type I IFN activity was associated with endothelial activation, measured by high sVCAM-1 (OR 1.68, p<0.01) and elevated EMPs (OR 1.40, p=0.03). Patients with SLE with high type I IFN activity had lower RHI than HCs (OR 2.61, p=0.04), indicating endothelial dysfunction.Deposition of complement factors on platelets, a measure of platelet activation, was seen in patients with endothelial dysfunction. High levels of sVCAM-1 were associated with increased deposition of C4d (OR 4.57, p<0.01) and C1q (OR 4.10, p=0.04) on platelets. High levels of EMPs were associated with C4d deposition on platelets (OR 3.64, p=0.03). Conclusions Endothelial dysfunction was associated with activation of platelets and the type I IFN system. We suggest that an interplay between the type I IFN system, injured endothelium and activated platelets may contribute to development of CVD in SLE.
Collapse
Affiliation(s)
- Helena Tydén
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Christian Lood
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Birgitta Gullstrand
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Christoffer Tandrup Nielsen
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark.,Copenhagen Lupus and Vasculitis Clinic, Centre for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels H H Heegaard
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Robin Kahn
- Department of Pediatrics, Clinical Sciences Lund, Lunds Universitey, Lund, Sweden
| | - Andreas Jönsen
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| | - Anders A Bengtsson
- Department of Rheumatology, Clinical Sciences Lund, Lunds University, Lund, Sweden
| |
Collapse
|
44
|
Buie JJ, Renaud LL, Muise-Helmericks R, Oates JC. IFN-α Negatively Regulates the Expression of Endothelial Nitric Oxide Synthase and Nitric Oxide Production: Implications for Systemic Lupus Erythematosus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1979-1988. [PMID: 28779021 PMCID: PMC5587385 DOI: 10.4049/jimmunol.1600108] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/29/2017] [Indexed: 01/11/2023]
Abstract
Systemic lupus erythematosus (SLE) is a known risk factor for endothelial dysfunction. Murine and human lupus studies revealed a role for IFN-α in vascular abnormalities associated with impaired blood vessel dilation. However, the impact of IFN-α on mediators that induce vasodilation and modulate inflammation, including endothelial NO synthase (eNOS) and NO bioavailability, are unknown. The objectives of this study were to determine how IFN-α promotes endothelial dysfunction in SLE, focusing on its regulation of eNOS and NO production in endothelial cells. We demonstrate that IFN-α promotes an endothelial dysfunction signature in HUVECs that is characterized by transcription suppression and mRNA instability of eNOS complemented by upregulation of MCP1 and VCAM1 These changes are associated with IFN-inducible gene expression. IFN-α impairs insulin-mediated NO production, and altered gene expression resulted from eNOS instability, possibly due to enhanced miR-155 expression. IFN-α significantly impaired NO production in insulin-stimulated HUVECs. IFN-α treatment also led to enhanced neutrophil adhesion. Our study introduces a novel pathway by which IFN-α serves as a proatherogenic mediator through repression of eNOS-dependent pathways. This could promote the development of endothelial dysfunction and cardiovascular disease in SLE.
Collapse
Affiliation(s)
- Joy Jones Buie
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
- Department of Molecular and Cellular Biology and Pathobiology, Medical University of South Carolina, Charleston, SC 29425
| | - Ludivine L Renaud
- Department of Molecular and Cellular Biology and Pathobiology, Medical University of South Carolina, Charleston, SC 29425
| | - Robin Muise-Helmericks
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425; and
| | - Jim C Oates
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425;
- Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401
| |
Collapse
|
45
|
Furumoto Y, Smith CK, Blanco L, Zhao W, Brooks SR, Thacker SG, Abdalrahman Z, Sciumè G, Tsai WL, Trier AM, Nunez L, Mast L, Hoffmann V, Remaley AT, O'Shea JJ, Kaplan MJ, Gadina M. Tofacitinib Ameliorates Murine Lupus and Its Associated Vascular Dysfunction. Arthritis Rheumatol 2017; 69:148-160. [PMID: 27429362 DOI: 10.1002/art.39818] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 07/12/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Dysregulation of innate and adaptive immune responses contributes to the pathogenesis of systemic lupus erythematosus (SLE) and its associated premature vascular damage. No drug to date targets both systemic inflammatory disease and the cardiovascular complications of SLE. Tofacitinib is a JAK inhibitor that blocks signaling downstream of multiple cytokines implicated in lupus pathogenesis. While clinical trials have shown that tofacitinib exhibits significant clinical efficacy in various autoimmune diseases, its role in SLE and the associated vascular pathology remains to be characterized. METHODS MRL/lpr lupus-prone mice were administered tofacitinib or vehicle by gavage for 6 weeks (therapeutic arm) or 8 weeks (preventive arm). Nephritis, skin inflammation, serum levels of autoantibodies and cytokines, mononuclear cell phenotype and gene expression, neutrophil extracellular traps (NETs) release, endothelium-dependent vasorelaxation, and endothelial differentiation were compared in treated and untreated mice. RESULTS Treatment with tofacitinib led to significant improvement in measures of disease activity, including nephritis, skin inflammation, and autoantibody production. In addition, tofacitinib treatment reduced serum levels of proinflammatory cytokines and interferon responses in splenocytes and kidney tissue. Tofacitinib also modulated the formation of NETs and significantly increased endothelium-dependent vasorelaxation and endothelial differentiation. The drug was effective in both preventive and therapeutic strategies. CONCLUSION Tofacitinib modulates the innate and adaptive immune responses, ameliorates murine lupus, and improves vascular function. These results indicate that JAK inhibitors have the potential to be beneficial in SLE and its associated vascular damage.
Collapse
Affiliation(s)
- Yasuko Furumoto
- Translational Immunology Section, Office of Science Technology (OST), NIAMS, NIH
| | | | - Luz Blanco
- Systemic Autoimmunity Branch, NIAMS, NIH
| | - Wenpu Zhao
- Systemic Autoimmunity Branch, NIAMS, NIH
| | | | | | | | | | - Wanxia L Tsai
- Translational Immunology Section, Office of Science Technology (OST), NIAMS, NIH
| | - Anna M Trier
- Translational Immunology Section, Office of Science Technology (OST), NIAMS, NIH
| | - Leti Nunez
- Translational Immunology Section, Office of Science Technology (OST), NIAMS, NIH
| | - Laurel Mast
- Translational Immunology Section, Office of Science Technology (OST), NIAMS, NIH
| | - Victoria Hoffmann
- Diagnostic and Research Services Branch, Office of the Director, NIH
| | | | - John J O'Shea
- Molecular Immunology and Inflammation Branch, NIAMS, NIH
| | | | - Massimo Gadina
- Translational Immunology Section, Office of Science Technology (OST), NIAMS, NIH
| |
Collapse
|
46
|
Grenn RC, Yalavarthi S, Gandhi AA, Kazzaz NM, Núñez-Álvarez C, Hernández-Ramírez D, Cabral AR, McCune WJ, Bockenstedt PL, Knight JS. Endothelial progenitor dysfunction associates with a type I interferon signature in primary antiphospholipid syndrome. Ann Rheum Dis 2017; 76:450-457. [PMID: 27432357 PMCID: PMC5233467 DOI: 10.1136/annrheumdis-2016-209442] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/03/2016] [Accepted: 06/25/2016] [Indexed: 01/16/2023]
Abstract
OBJECTIVES Patients with antiphospholipid syndrome (APS) are at risk for subclinical endothelial injury, as well as accelerated atherosclerosis. In the related disease systemic lupus erythematosus, there is a well-established defect in circulating endothelial progenitors, which leads to an accrual of endothelial damage over time. This defect has been at least partially attributed to exaggerated expression of type I interferons (IFNs). We sought to determine whether these pathways are important in APS. METHODS We studied 68 patients with primary APS. Endothelial progenitors were assessed by flow cytometry and functional assay. Type I IFN activity was determined by a well-accepted bioassay, while peripheral blood mononuclear cells were scored for expression of IFN-responsive genes. RESULTS Endothelial progenitors from patients with APS demonstrated a marked defect in the ability to differentiate into endothelial cells, a phenotype which could be mimicked by treating control progenitors with APS sera. Elevated type I IFN activity was detected in the circulation of patients with APS (a finding that was then replicated in an independent cohort). While IgG depletion from APS sera did not rescue endothelial progenitor function, the dysfunction was successfully reversed by a type I IFN receptor-neutralising antibody. CONCLUSIONS We describe, for the first time to our knowledge, an IFN signature in primary APS and show that this promotes impaired endothelial progenitor function. This work opens the door to novel approaches that may mitigate vascular damage in APS, such as anti-IFN drugs.
Collapse
Affiliation(s)
- Robert C. Grenn
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alex A. Gandhi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nayef M. Kazzaz
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Carlos Núñez-Álvarez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diego Hernández-Ramírez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Antonio R. Cabral
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - W. Joseph McCune
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Paula L. Bockenstedt
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
47
|
Kröger W, Mapiye D, Entfellner JBD, Tiffin N. A meta-analysis of public microarray data identifies gene regulatory pathways deregulated in peripheral blood mononuclear cells from individuals with Systemic Lupus Erythematosus compared to those without. BMC Med Genomics 2016; 9:66. [PMID: 27846842 PMCID: PMC5111272 DOI: 10.1186/s12920-016-0227-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 10/21/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Systemic Lupus Erythematosus (SLE) is a complex, multi-systemic, autoimmune disease for which the underlying aetiological mechanisms are poorly understood. The genetic and molecular processes underlying lupus have been extensively investigated using a variety of -omics approaches, including genome-wide association studies, candidate gene studies and microarray experiments of differential gene expression in lupus samples compared to controls. METHODS This study analyses a combination of existing microarray data sets to identify differentially regulated genetic pathways that are dysregulated in human peripheral blood mononuclear cells from SLE patients compared to unaffected controls. Two statistical approaches, quantile discretisation and scaling, are used to combine publicly available expression microarray datasets and perform a meta-analysis of differentially expressed genes. RESULTS Differentially expressed genes implicated in interferon signaling were identified by the meta-analysis, in agreement with the findings of the individual studies that generated the datasets used. In contrast to the individual studies, however, the meta-analysis and subsequent pathway analysis additionally highlighted TLR signaling, oxidative phosphorylation and diapedesis and adhesion regulatory networks as being differentially regulated in peripheral blood mononuclear cells (PBMCs) from SLE patients compared to controls. CONCLUSION Our analysis demonstrates that it is possible to derive additional information from publicly available expression data using meta-analysis techniques, which is particularly relevant to research into rare diseases where sample numbers can be limiting.
Collapse
Affiliation(s)
- Wendy Kröger
- South African National Bioinformatics Institute/Medical Research Council of South Africa Bioinformatics Capacity Development Unit, University of the Western Cape, Cape Town, South Africa
| | - Darlington Mapiye
- South African National Bioinformatics Institute/Medical Research Council of South Africa Bioinformatics Capacity Development Unit, University of the Western Cape, Cape Town, South Africa
| | - Jean-Baka Domelevo Entfellner
- South African National Bioinformatics Institute/Medical Research Council of South Africa Bioinformatics Capacity Development Unit, University of the Western Cape, Cape Town, South Africa
| | - Nicki Tiffin
- South African National Bioinformatics Institute/Medical Research Council of South Africa Bioinformatics Capacity Development Unit, University of the Western Cape, Cape Town, South Africa
| |
Collapse
|
48
|
Relation of peritubular capillary features to class of lupus nephritis. BMC Nephrol 2016; 17:169. [PMID: 27829387 PMCID: PMC5103390 DOI: 10.1186/s12882-016-0388-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/03/2016] [Indexed: 02/05/2023] Open
Abstract
Background Experimental studies have linked peritubular capillary (PTC) loss with progression of chronic kidney disease. Minimal information on PTC in lupus nephritis (LN) has been reported. We therefore evaluated the PTC area in different classes of LN and determined if specific clinical characteristics correlated with PTC changes. Methods Renal biopsies of 253 subjects with LN (categorized using the ISN/RPS 2003 classification) and 13 normal renal donors (the controls) were retrospectively evaluated for PTC morphology by staining for CD31 with immunohistochemistry method. The percent positive area of PTC (% PTC) was correlated with serum and urinary measures of renal function and renal pathology. Results Significant PTC loss was observed in all classes of LN compared to controls. The % PTC area was highest in controls (7.64±1.48 %) with levels of 1.95±1.50, 4.16±3.85, 4.19±4.45, 5.02±1.79, and 4.45±3.75 in classes II, III, IV, IV combined with V and V, respectively (all p values < 0.05). The lowest PTC density was observed in class II LN, but this may be because some cases with worse classes of LN showed increased PTC density due to abnormally dilated capillaries associated with acute inflammation and angiogenesis. %PTC was increased in those with hematuria (5.8±5.2 vs. 3.6±3.4 %, red blood cells 3-10 vs. < 3 cells/high power field, p < 0.05) and was reduced in those with a moderately declined renal function (3.29±3.40 vs. 4.42±4.12, eGFR 15-59 vs. ≥ 60 ml/min/1.73 m2, p < 0.05). Nephrotic-range proteinuria also trended to be associated with lower PTC density although it did not reach statistical significance (3.1±2.6 vs. 4.9±4.5, p= 0.067). Conclusions LN is associated with PTC loss and the severity correlates with reduced renal function. Further studies are needed to investigate whether a loss of PTC can predict long term renal outcomes in LN. Electronic supplementary material The online version of this article (doi:10.1186/s12882-016-0388-2) contains supplementary material, which is available to authorized users.
Collapse
|
49
|
Abstract
Chronic kidney disease (CKD) is a lethal and rapidly increasing burden on society. Despite this, there are relatively few therapies in development for the treatment of CKD. Several recent costly phase 3 trials have failed to provide improved renal outcomes, diminishing interest in pharmaceutical investment. Furthermore, poor patient, physician, and payer awareness of CKD as a diagnosis has contributed to slow trial enrollment and successful implementation of these trials. Nevertheless, several therapeutics remain in development for the treatment of CKD, including mineralocorticoid-receptor antagonists, sodium/glucose cotransporter 2 inhibitors, anti-inflammatory drugs, and drugs that mitigate oxidative injury. Success of future CKD therapeutic trials will depend not only on improved understanding of disease pathogenesis, but also on improved trial enrollment rates, through increasing awareness of this disease by the public, policy makers, and the greater medical community.
Collapse
Affiliation(s)
- Matthew D Breyer
- Biotechnology Discovery Research, Eli Lilly and Company, Indianapolis, IN.
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
50
|
Lennard Richard ML, Brandon D, Lou N, Sato S, Caldwell T, Nowling TK, Gilkeson G, Zhang XK. Acetylation impacts Fli-1-driven regulation of granulocyte colony stimulating factor. Eur J Immunol 2016; 46:2322-2332. [PMID: 27431361 DOI: 10.1002/eji.201646315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/22/2016] [Accepted: 07/15/2016] [Indexed: 11/07/2022]
Abstract
Fli-1 has emerged as a critical regulator of inflammatory mediators, including MCP-1, CCL5, and IL-6. The cytokine, granulocyte colony stimulating factor (G-CSF) regulates neutrophil precursor maturation and survival, and activates mature neutrophils. Previously, a significant decrease in neutrophil infiltration into the kidneys of Fli-1+/- lupus-prone mice was observed. In this study, a significant decrease in G-CSF protein expression was detected in stimulated murine and human endothelial cells when expression of Fli-1 was inhibited. The murine G-CSF promoter contains numerous putative Fli-1 binding sites and several regions within the proximal promoter are significantly enriched for Fli-1 binding. Transient transfection assays indicate that Fli-1 drives transcription from the G-CSF promoter and mutation of the Fli-1 DNA binding domain resulted in a 94% loss of transcriptional activation. Mutation of a known acetylation site, led to a significant increase in G-CSF promoter activation. The histone acetyltransferases p300/CBP and p300/CBP associated factor (PCAF) significantly decrease Fli-1 specific activation of the G-CSF promoter. Thus, acetylation appears to be an important mechanism behind Fli-1 driven activation of the G-CSF promoter. These results further support the theory that Fli-1 plays a major role in the regulation of several inflammatory mediators, ultimately affecting inflammatory disease pathogenesis.
Collapse
Affiliation(s)
- Mara L Lennard Richard
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Danielle Brandon
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Ning Lou
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA.,Jinan Central Hospital, Shandong University, Jinan, Shangdong, China
| | - Shuzo Sato
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Tomika Caldwell
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Tamara K Nowling
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Gary Gilkeson
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA.,Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA
| | - Xian K Zhang
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA. .,Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|