1
|
Lu Z, Ni W, Wu Y, Zhai B, Zhao Q, Zheng T, Liu Q, Ding D. Application of biomarkers in the diagnosis of kidney disease. Front Med (Lausanne) 2025; 12:1560222. [PMID: 40370722 PMCID: PMC12075424 DOI: 10.3389/fmed.2025.1560222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Worldwide, kidney disease has grown to be an important global public health agenda that reduces longevity. Medical institutions around the globe should enhance screening efforts for kidney disease, to facilitate early kidney disease detection, diagnosis, and intervention. Common screening methods for nephropathy encompass renal tissue biopsy, urine dry chemistry tests, urine formed element analysis, and urine-specific protein assays, among others. These methodologies evaluate renal health by scrutinizing a spectrum of biomarkers. Precise classification and quantitative analysis of these biomarkers can assist in determining the site and extent of kidney injury, as well as in assessing treatment efficacy and prognosis. In this paper, we reviewed the methods and biomarkers for kidney disease and also the integration of multiple biomarkers. With the aim of reasonable applying these markers to the early detection, accurate diagnosis, and scientific management of kidney disease, thereby mitigating the threat posed by kidney disease to human health.
Collapse
Affiliation(s)
- Zuohua Lu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Weifeng Ni
- Department of Endocrinology, Rheumatology and Immunology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yuding Wu
- Goldsite Diagnostics Inc., Shenzhen, China
| | - Bin Zhai
- Department of Clinical Laboratory, Baotou Central Hospital, Baotou, China
| | - Qiuyun Zhao
- Department of Clinical Laboratory, Guilin Hospital of Integrated Traditional Chinese and Western Medicine, Guilin, China
| | - Tian Zheng
- Department of Clinical Laboratory, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qianqian Liu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Dapeng Ding
- Department of Clinical Laboratory, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
2
|
Jiang K, Pan Y, Pu D, Shi L, Xu X, Bai M, Gong X, Guo J, Li M. Kidney transplantation in Lupus Nephritis: a comprehensive review of challenges and strategies. BMC Surg 2025; 25:112. [PMID: 40121458 PMCID: PMC11929324 DOI: 10.1186/s12893-025-02832-w] [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: 11/20/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
PURPOSE OF REVIEW: Lupus nephritis (LN) is a severe complication of systemic lupus erythematosus (SLE), significantly impacting patient outcomes. Despite advances in immunosuppressive therapies, many patients progress to end-stage renal disease (ESRD), and kidney transplantation becomes essential for improving survival. However, the unique characteristics of autoimmune diseases make the timing of kidney transplantation and post-transplant management challenging. This review evaluates authoritative guidelines and recent studies to identify optimal timing for kidney transplantation and effective pre- and post-transplant management measures for patients with LN. RECENT FINDINGS: Advancements in immunosuppressive therapies, including calcineurin inhibitors, Voclosporin, and biologic agents such as belimumab, have significantly improved LN management. Emerging biomarkers, such as urinary MCP-1 and BAFF, offer promising tools for monitoring LN activity and predicting recurrence risk post-transplantation. Current guidelines emphasize the importance of achieving disease quiescence before transplantation, while new evidence supports the benefits of preemptive transplantation and personalized immunosuppressive regimens in improving patient and graft survival. This review highlights the latest evidence and strategies for optimizing kidney transplantation outcomes in LN patients, focusing on timing, immunosuppression, and disease monitoring.
Collapse
Affiliation(s)
- Kerong Jiang
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China.
| | - Yongsheng Pan
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Dan Pu
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Lijuan Shi
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Xiaoliang Xu
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Minfeng Bai
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Xiaqiong Gong
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Jie Guo
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Ming Li
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| |
Collapse
|
3
|
Saegusa K, Tsuchida Y, Komai T, Tsuchiya H, Fujio K. Advances in Targeted Therapy for Systemic Lupus Erythematosus: Current Treatments and Novel Approaches. Int J Mol Sci 2025; 26:929. [PMID: 39940698 PMCID: PMC11816971 DOI: 10.3390/ijms26030929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with diverse clinical manifestations that can lead to severe organ damage. The complex pathophysiology of SLE makes treatment selection difficult. This review examines the current evidence for biological therapies in SLE, including the anti-B cell activating factor antibody belimumab; the type I interferon receptor antagonist anifrolumab; the novel calcineurin inhibitor voclosporin; and rituximab, which targets CD20 on B cells. We also describe emerging therapies, including novel agents in development and CD19-directed chimeric antigen receptor (CAR) T cell therapy, which has shown promise in early clinical experience. Recent advances in biomarker research, including interferon signatures and transcriptomic profiles, may facilitate patient stratification and treatment selection. This review offers insights into current and future treatment strategies for patients with SLE by analyzing clinical trial results and recent immunological findings.
Collapse
Affiliation(s)
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; (K.S.); (T.K.); (H.T.); (K.F.)
| | | | | | | |
Collapse
|
4
|
Bhoj PS, Nocito C, Togre NS, Winfield M, Lubinsky C, Khan S, Mogadala N, Seliga A, Unterwald EM, Persidsky Y, Sriram U. Tissue Kallikrein-1 Suppresses Type I Interferon Responses and Reduces Depressive-Like Behavior in the MRL/lpr Lupus-Prone Mouse Model. Int J Mol Sci 2024; 25:10080. [PMID: 39337564 PMCID: PMC11432477 DOI: 10.3390/ijms251810080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Excessive production and response to Type I interferons (IFNs) is a hallmark of systemic lupus erythematosus (SLE). Neuropsychiatric lupus (NPSLE) is a common manifestation of human SLE, with major depression as the most common presentation. Clinical studies have demonstrated that IFNα can cause depressive symptoms. We have shown that the kallikrein-kinin system (KKS) [comprised of kallikreins (Klks) and bradykinins] and angiotensin-converting enzyme inhibitors suppressed Type I IFN responses in dendritic cells from lupus-prone mice and human peripheral blood mononuclear cells. Tissue Klk genes are decreased in patients with lupus, and giving exogenous Klk1 ameliorated kidney pathology in mice. We retro-orbitally administered mouse klk1 gene-carrying adenovirus in the Murphy Roths Large lymphoproliferative (MRL/lpr) lupus-prone mice at early disease onset and analyzed immune responses and depressive-like behavior. Klk1 improved depressive-like behavior, suppressed interferon-responsive genes and neuroinflammation, and reduced plasma IFNα levels and proinflammatory cytokines. Klk1 also reduced IFNAR1 and JAK1 protein expression, important upstream molecules in Type I IFN signaling. Klk1 reduced bradykinin B1 receptor expression, which is known to induce proinflammatory response. Together, these findings suggest that Klk1 may be a potential therapeutic candidate to control IFNα production/responses and other inflammatory responses in SLE and NPSLE.
Collapse
Affiliation(s)
- Priyanka S. Bhoj
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Cassandra Nocito
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Namdev S. Togre
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Malika Winfield
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Cody Lubinsky
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Sabeeya Khan
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Nikhita Mogadala
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Alecia Seliga
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Ellen M. Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Uma Sriram
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| |
Collapse
|
5
|
Guo Q, Qiao P, Wang J, Zhao L, Guo Z, Li X, Fan X, Yu C, Zhang L. Investigating the value of urinary biomarkers in relation to lupus nephritis histopathology: present insights and future prospects. Front Pharmacol 2024; 15:1421657. [PMID: 39104393 PMCID: PMC11298450 DOI: 10.3389/fphar.2024.1421657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Lupus nephritis (LN), a leading cause of death in Systemic Lupus Erythematosus (SLE) patients, presents significant diagnostic and prognostic challenges. Although renal pathology offers critical insights regarding the diagnosis, classification, and therapy for LN, its clinical utility is constrained by the invasive nature and limited reproducibility of renal biopsies. Moreover, the continuous monitoring of renal pathological changes through repeated biopsies is impractical. Consequently, there is a growing interest in exploring urine as a non-invasive, easily accessible, and dynamic "liquid biopsy" alternative to guide clinical management. This paper examines novel urinary biomarkers from a renal pathology perspective, encompassing cellular components, cytokines, adhesion molecules, auto-antibodies, soluble leukocyte markers, light chain fragments, proteins, small-molecule peptides, metabolomics, urinary exosomes, and ribonucleic acids. We also discuss the application of combined models comprising multiple biomarkers in assessing lupus activity. These innovative biomarkers and models offer insights into LN disease activity, acute and chronic renal indices, fibrosis, thrombotic microangiopathy, podocyte injury, and other pathological changes, potentially improving the diagnosis, management, and prognosis of LN. These urinary biomarkers or combined models may serve as viable alternatives to traditional renal pathology, potentially revolutionizing the method for future LN diagnosis and observation.
Collapse
Affiliation(s)
- Qianyu Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Pengyan Qiao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Juanjuan Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Stem Cell Translational Laboratory, Shanxi Bethune Hospital, Taiyuan, China
| | - Li Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Zhiying Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Xiaochen Li
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Xiuying Fan
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Office of Drug Clinical Trial Institution, Taiyuan, China
| | - Chong Yu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Liyun Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
- Stem Cell Translational Laboratory, Shanxi Bethune Hospital, Taiyuan, China
- Office of Drug Clinical Trial Institution, Taiyuan, China
| |
Collapse
|
6
|
Lemmon B, Kyrgiou M, Mullins E, Khullar V. Cytokines in Bladder Pain Syndrome: A Review of the Literature. Int Urogynecol J 2024; 35:1119-1129. [PMID: 38771505 DOI: 10.1007/s00192-024-05778-4] [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: 10/26/2023] [Accepted: 01/31/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION AND HYPOTHESIS Bladder pain syndrome (BPS) is poorly understood with both the aetiology and pathophysiology being unknown. Symptoms overlap with other disorders, such as overactive bladder (OAB) and chronic pelvic pain disorders such as endometriosis, making a consensus on how to diagnosis and manage patients challenging. The development of biomarkers for BPS may be the key to understanding more about its pathophysiology, as well as aiding diagnosis, subclassification, and discovering new drug targets for its management. As inflammation is widely understood to hold a central role in BPS, the evaluation of cytokines has gained interest. This article summarises the current literature and understanding of urinary, serum, and bladder tissue cytokines found elevated in patients with bladder pain syndrome. METHODS literature search using Pub Med with the keywords "bladder pain syndrome", "painful bladder syndrome", "bladder pain", "Interstitial cystitis" AND "cytokines" or "inflammation". This study was except from institutional approval. RESULTS Thirty-six cytokines have been identified as being statistically significantly elevated in either the serum, urine, or bladder tissue of patients with bladder pain syndrome in the 22 studies identified in this review of the literature. These cytokines include those from the interleukin group (n = 14), the CXC chemokine group (n = 5), and the C-C chemokine group (n = 7). CONCLUSIONS CXCL-1, CXCL-8, CXCL-9, CXCL-10, CXCL-11 from the CXC chemokine group, and CCL2, CCL4, CCL5, CCL7, and CCL11 from the C-C chemokine group have been found to be significantly elevated in patients with bladder pain in the literature. Many of these analytes also have supporting evidence for their roles in bladder pain from animal models and studies in other chronic inflammatory conditions. It is likely that a single cytokine will not serve as an adequate biomarker of disease in bladder pain syndrome for either diagnosis or disease severity. Instead, panels of inflammatory mediators may reveal more about the different pathways of inflammation leading to similar presentations of bladder pain in patients.
Collapse
Affiliation(s)
- Bernadette Lemmon
- Urogynaecology Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, Praed Street, London, W2 1NY, UK.
| | - Maria Kyrgiou
- IRDB, Department of Metabolism, Digestion and Reproduction-Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Edward Mullins
- Urogynaecology Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, Praed Street, London, W2 1NY, UK
| | - Vikram Khullar
- Urogynaecology Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, Praed Street, London, W2 1NY, UK
| |
Collapse
|
7
|
Martinez Valenzuela L, Gómez-Preciado F, Guiteras J, Antón Pampols P, Gomà M, Fulladosa X, Cruzado JM, Torras J, Draibe J. Immune checkpoint inhibitors induce acute interstitial nephritis in mice with increased urinary MCP1 and PD-1 glomerular expression. J Transl Med 2024; 22:421. [PMID: 38702780 PMCID: PMC11069287 DOI: 10.1186/s12967-024-05177-9] [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: 02/05/2024] [Accepted: 04/05/2024] [Indexed: 05/06/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) induce acute interstitial nephritis (AIN) in 2-5% of patients, with a clearly higher incidence when they are combined with platinum derivatives. Unfortunately, suitable disease models and non-invasive biomarkers are lacking. To fill this gap in our understanding, we investigated the renal effects of cisplatin and anti-PD-L1 antibodies in mice, assessing PD-1 renal expression and cytokine levels in mice with AIN, and then we compared these findings with those in AIN-diagnosed cancer patients. METHODS Twenty C57BL6J mice received 200 µg of anti-PD-L1 antibody and 5 mg/kg cisplatin intraperitoneally and were compared with those receiving cisplatin (n = 6), anti-PD-L1 (n = 7), or saline (n = 6). After 7 days, the mice were euthanized. Serum and urinary concentrations of TNFα, CXCL10, IL-6, and MCP-1 were measured by Luminex. The kidney sections were stained to determine PD-1 tissue expression. Thirty-nine cancer patients with AKI were enrolled (AIN n = 33, acute tubular necrosis (ATN) n = 6), urine MCP-1 (uMCP-1) was measured, and kidney sections were stained to assess PD-1 expression. RESULTS Cisplatin and anti PD-L1 treatment led to 40% AIN development (p = 0.03) in mice, accompanied by elevated serum creatinine and uMCP1. AIN-diagnosed cancer patients also had higher uMCP1 levels than ATN-diagnosed patients, confirming our previous findings. Mice with AIN exhibited interstitial PD-1 staining and stronger glomerular PD-1 expression, especially with combination treatment. Conversely, human AIN patients only showed interstitial PD-1 positivity. CONCLUSIONS Only mice receiving cisplatin and anti-PDL1 concomitantly developed AIN, accompanied with a more severe kidney injury. AIN induced by this drug combination was linked to elevated uMCP1, consistently with human AIN, suggesting that uMCP1 can be potentially used as an AIN biomarker.
Collapse
Affiliation(s)
- Laura Martinez Valenzuela
- Nephrology Department, Bellvitge University Hospital. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Feixa Llarga S/N, Barcelona, 08907, Spain.
| | - Francisco Gómez-Preciado
- Nephrology Department, Bellvitge University Hospital. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Feixa Llarga S/N, Barcelona, 08907, Spain
| | - Jordi Guiteras
- Experimental Nephrology Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, 08907, Spain
- Fundació Bosch i Gimpera, University of Barcelona, Barcelona, 08028, Spain
| | - Paula Antón Pampols
- Nephrology Department, Bellvitge University Hospital. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Feixa Llarga S/N, Barcelona, 08907, Spain
| | - Montserrat Gomà
- Pathology Department, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, 08907, Spain
| | - Xavier Fulladosa
- Nephrology Department, Bellvitge University Hospital. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Feixa Llarga S/N, Barcelona, 08907, Spain
- Faculty of Medicine, Bellvitge Campus, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, 08907, Spain
| | - Josep Maria Cruzado
- Nephrology Department, Bellvitge University Hospital. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Feixa Llarga S/N, Barcelona, 08907, Spain
- Faculty of Medicine, Bellvitge Campus, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, 08907, Spain
| | - Joan Torras
- Nephrology Department, Bellvitge University Hospital. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Feixa Llarga S/N, Barcelona, 08907, Spain
- Faculty of Medicine, Bellvitge Campus, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, 08907, Spain
| | - Juliana Draibe
- Nephrology Department, Bellvitge University Hospital. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Feixa Llarga S/N, Barcelona, 08907, Spain
| |
Collapse
|
8
|
Liu TT, Sun HF, Han YX, Zhan Y, Jiang JD. The role of inflammation in silicosis. Front Pharmacol 2024; 15:1362509. [PMID: 38515835 PMCID: PMC10955140 DOI: 10.3389/fphar.2024.1362509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Silicosis is a chronic illness marked by diffuse fibrosis in lung tissue resulting from continuous exposure to SiO2-rich dust in the workplace. The onset and progression of silicosis is a complicated and poorly understood pathological process involving numerous cells and molecules. However, silicosis poses a severe threat to public health in developing countries, where it is the most prevalent occupational disease. There is convincing evidence supporting that innate and adaptive immune cells, as well as their cytokines, play a significant role in the development of silicosis. In this review, we describe the roles of immune cells and cytokines in silicosis, and summarize current knowledge on several important inflammatory signaling pathways associated with the disease, aiming to provide novel targets and strategies for the treatment of silicosis-related inflammation.
Collapse
Affiliation(s)
| | | | | | - Yun Zhan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | |
Collapse
|
9
|
Gholamalizadeh H, Ensan B, Sukhorukov VN, Sahebkar A. Targeting the CCL2-CCR2 signaling pathway: potential implications of statins beyond cardiovascular diseases. J Pharm Pharmacol 2024; 76:138-153. [PMID: 38127312 DOI: 10.1093/jpp/rgad112] [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: 07/13/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND The chemokine ligand CCL2 and its cognate receptor CCR2 have been implicated in the pathogenesis of a wide variety of diseases. Hence, the inhibition of the CCL2/CCR2 signaling pathway has been of great attention in recent studies. Among suggested medications, statins known as HMG-COA reductase inhibitors with their pleiotropic effects are widely under investigation. METHOD A comprehensive literature search on Scopus and PubMed databases was conducted using the keywords 'CCL2', 'CCR2', 'monocyte chemoattractant protein-1', 'HMG-COA reductase inhibitor', and 'statin'. Both experimental and clinical studies measuring CCL2/CCR2 expressions following statin therapy were identified excluding the ones focused on cardiovascular diseases. RESULTS Herein, we summarized the effects of statins on CCL2 and CCR2 expression in various pathologic conditions including immune-mediated diseases, nephropathies, diabetes, rheumatic diseases, neuroinflammation, inflammatory bowel diseases, gynecologic diseases, and cancers. CONCLUSION For the most part, statins play an inhibitory role on the CCL2-CCR2 axis which implies their potential to be further developed as therapeutic options in non-cardiovascular diseases either alone or in combination with other conventional treatments. However, the existing literature mostly focused on experimental models and is therefore inadequate to reach a conclusion.
Collapse
Affiliation(s)
- Hanieh Gholamalizadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Behzad Ensan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow 125315, Russia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| |
Collapse
|
10
|
Nourie N, Ghaleb R, Lefaucheur C, Louis K. Toward Precision Medicine: Exploring the Landscape of Biomarkers in Acute Kidney Injury. Biomolecules 2024; 14:82. [PMID: 38254682 PMCID: PMC10813773 DOI: 10.3390/biom14010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Acute kidney injury (AKI) remains a complex challenge with diverse underlying pathological mechanisms and etiologies. Current detection methods predominantly rely on serum creatinine, which exhibits substantial limitations in specificity and poses the issue of late-stage detection of kidney injury. In this review, we propose an up-to-date and comprehensive summary of advancements that identified novel biomarker candidates in blood and urine and ideal criteria for AKI biomarkers such as renal injury specificity, mechanistic insight, prognostic capacity, and affordability. Recently identified biomarkers not only indicate injury location but also offer valuable insights into a range of pathological processes, encompassing reduced glomerular filtration rate, tubular function, inflammation, and adaptive response to injury. The clinical applications of AKI biomarkers are becoming extensive and serving as relevant tools in distinguishing acute tubular necrosis from other acute renal conditions. Also, these biomarkers can offer significant insights into the risk of progression to chronic kidney disease CKD and in the context of kidney transplantation. Integration of these biomarkers into clinical practice has the potential to improve early diagnosis of AKI and revolutionize the design of clinical trials, offering valuable endpoints for therapeutic interventions and enhancing patient care and outcomes.
Collapse
Affiliation(s)
- Nicole Nourie
- Department of Nephrology and Kidney Transplantation, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
- Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, 75010 Paris, France
| | - Rita Ghaleb
- Faculty of Medicine, Saint Joseph University, Beirut 1104 2020, Lebanon
| | - Carmen Lefaucheur
- Department of Nephrology and Kidney Transplantation, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
- Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, 75010 Paris, France
| | - Kevin Louis
- Department of Nephrology and Kidney Transplantation, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
- Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, 75010 Paris, France
| |
Collapse
|
11
|
Rogers ML, Schultz DW, Karnaros V, Shepheard SR. Urinary biomarkers for amyotrophic lateral sclerosis: candidates, opportunities and considerations. Brain Commun 2023; 5:fcad287. [PMID: 37946793 PMCID: PMC10631861 DOI: 10.1093/braincomms/fcad287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/23/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Amyotrophic lateral sclerosis is a relentless neurodegenerative disease that is mostly fatal within 3-5 years and is diagnosed on evidence of progressive upper and lower motor neuron degeneration. Around 15% of those with amyotrophic lateral sclerosis also have frontotemporal degeneration, and gene mutations account for ∼10%. Amyotrophic lateral sclerosis is a variable heterogeneous disease, and it is becoming increasingly clear that numerous different disease processes culminate in the final degeneration of motor neurons. There is a profound need to clearly articulate and measure pathological process that occurs. Such information is needed to tailor treatments to individuals with amyotrophic lateral sclerosis according to an individual's pathological fingerprint. For new candidate therapies, there is also a need for methods to select patients according to expected treatment outcomes and measure the success, or not, of treatments. Biomarkers are essential tools to fulfil these needs, and urine is a rich source for candidate biofluid biomarkers. This review will describe promising candidate urinary biomarkers of amyotrophic lateral sclerosis and other possible urinary candidates in future areas of investigation as well as the limitations of urinary biomarkers.
Collapse
Affiliation(s)
- Mary-Louise Rogers
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia
| | - David W Schultz
- Neurology Department and MND Clinic, Flinders Medical Centre, Adelaide 5042, South Australia, Australia
| | - Vassilios Karnaros
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia
| | - Stephanie R Shepheard
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia
| |
Collapse
|
12
|
Fasano S, Milone A, Nicoletti GF, Isenberg DA, Ciccia F. Precision medicine in systemic lupus erythematosus. Nat Rev Rheumatol 2023; 19:331-342. [PMID: 37041269 DOI: 10.1038/s41584-023-00948-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/13/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that has diverse clinical manifestations, ranging from restricted cutaneous involvement to life-threatening systemic organ involvement. The heterogeneity of pathomechanisms that lead to SLE contributes to between-patient variation in clinical phenotype and treatment response. Ongoing efforts to dissect cellular and molecular heterogeneity in SLE could facilitate the future development of stratified treatment recommendations and precision medicine, which is a considerable challenge for SLE. In particular, some genes involved in the clinical heterogeneity of SLE and some phenotype-related loci (STAT4, IRF5, PDGF genes, HAS2, ITGAM and SLC5A11) have an association with clinical features of the disease. An important part is also played by epigenetic varation (in DNA methylation, histone modifications and microRNAs) that influences gene expression and affects cell function without modifying the genome sequence. Immune profiling can help to identify an individual's specific response to a therapy and can potentially predict outcomes, using techniques such as flow cytometry, mass cytometry, transcriptomics, microarray analysis and single-cell RNA sequencing. Furthermore, the identification of novel serum and urinary biomarkers would enable the stratification of patients according to predictions of long-term outcomes and assessments of potential response to therapy.
Collapse
Affiliation(s)
- Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| | - Alessandra Milone
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - David A Isenberg
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
13
|
Diagnostic test accuracy of novel biomarkers for lupus nephritis-An overview of systematic reviews. PLoS One 2022; 17:e0275016. [PMID: 36215243 PMCID: PMC9550089 DOI: 10.1371/journal.pone.0275016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with multiorgan inflammatory involvement and a mortality rate that is 2.6-fold higher than individuals of the same age and sex in the general population. Approximately 50% of patients with SLE develop renal impairment (lupus nephritis). Delayed diagnosis of lupus nephritis is associated with a higher risk of progression to end-stage renal disease, the need for replacement therapy, and mortality. The initial clinical manifestations of lupus nephritis are often discrete or absent and are usually detected through complementary tests. Although widely used in clinical practice, their accuracy is limited. A great scientific effort has been exerted towards searching for new, more sensitive, and specific biomarkers in recent years. Some systematic reviews have individually evaluated new serum and urinary biomarkers tested in patients with lupus nephritis. This overview aimed to summarize systematic reviews on the accuracy of novel serum and urinary biomarkers for diagnosing lupus nephritis in patients with SLE, discussing how our results can guide the clinical management of the disease and the direction of research in this area. METHODS The research question is "What is the accuracy of the new serum and urinary biomarkers studied for the diagnosis of LN in patients with SLE?". We searched for systematic reviews of observational studies evaluating the diagnostic accuracy of new serum or urinary biomarkers of lupus nephritis. The following databases were included: PubMed, EMBASE, BIREME/LILACS, Scopus, Web of Science, and Cochrane, including gray literature found via Google Scholar and PROQUEST. Two authors assessed the reviews for inclusion, data extraction, and assessment of the risk of bias (ROBIS tool). RESULTS Ten SRs on the diagnostic accuracy of new serum and urinary BMs in LN were selected. The SRs evaluated 7 distinct BMs: (a) antibodies (anti-Sm, anti-RNP, and anti-C1q), (b) cytokines (TWEAK and MCP-1), (c) a chemokine (IP-10), and (d) an acute phase glycoprotein (NGAL), in a total of 20 review arms (9 that analyzed serum BMs, and 12 that analyzed BMs in urine). The population evaluated in the primary studies was predominantly adults. Two SRs included strictly adults, 5 reviews also included studies in the paediatric population, and 4 did not report the age groups. The results of the evaluation with the ROBIS tool showed that most of the reviews had a low overall risk of bias. CONCLUSIONS There are 10 SRs of evidence relating to the diagnostic accuracy of serum and urinary biomarkers for lupus nephritis. Among the BMs evaluated, anti-C1q, urinary MCP-1, TWEAK, and NGAL stood out, highlighting the need for additional research, especially on LN diagnostic panels, and attempting to address methodological issues within diagnostic accuracy research. This would allow for a better understanding of their usefulness and possibly validate their clinical use in the future. REGISTRATION This project is registered on the International Prospective Registry of Systematic Reviews (PROSPERO) database (CRD42020196693).
Collapse
|
14
|
Li Y, Ding T, Chen J, Ji J, Wang W, Ding B, Ge W, Fan Y, Xu L. The protective capability of Hedyotis diffusa Willd on lupus nephritis by attenuating the IL-17 expression in MRL/lpr mice. Front Immunol 2022; 13:943827. [PMID: 35958622 PMCID: PMC9359319 DOI: 10.3389/fimmu.2022.943827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/28/2022] [Indexed: 12/04/2022] Open
Abstract
Lupus nephritis (LN), the most severe organ manifestation of systemic lupus erythematosus (SLE), is generally treated with glucocorticoids (GC) in clinical practice, leading to drug resistance and adverse effects in the long term. Fortunately, the combination of GC and traditional Chinese medical prescriptions can attenuate the adverse effects and improve therapeutic efficiency. Hedyotis diffusa Willd (HDW) is one of the most commonly used herbal compounds for LN treatment, which exhibits “heat-clearing” and “detoxification” effects. However, the underlying pharmacological mechanism remains unclear. The present study identified the chemical compounds in HDW extract with UPLC-Q-TOF-MS/MS. A total of 49 components were identified in the HDW extract, and the IL-17 signaling pathway was highly enriched by network pharmacological analysis. MRL/lpr model mice, reflecting the spontaneous development of LN, were used to evaluate the protective activity and investigate the underlying mechanism of the combination treatment. The white blood cell content (WBC), including lymphocytes and neutrophils, cytokines (IL-6, MCP-1, TNF-a), and various autoantibodies (ANA, ab-dsDNA, ab-snRNP/sm) in the blood of MRL/lpr mice were significantly improved by the intragastric administration of HDW. Additionally, the expression of STAT3, IL-17, Ly6G, and MPO in the kidney and neutrophil NETosis were ameliorated with HDW treatment. The pathological and morphological analysis suggested that HDW application could reduce urinary protein levels and inflammatory cell infiltration and inhibit glomerular interstitial cell proliferation. Hence, HDW might ameliorate lupus nephritis by inhibiting IL-6 secretion and STAT3-induced IL-17 expression. The active compounds in HDW were predictively selected with computational methods. The docking affinity of asiatic acid, neoandrographolide to IL-6, glycyrrhetinic acid, oleanolic acid, ursolic acid, and wilforlide A to STAT3 are extremely high. In conclusion, the IL-6 and STAT3/IL-17signaling pathways could be critical regulative targets of HDW on LN.
Collapse
Affiliation(s)
- Ying Li
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tao Ding
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinjun Ji
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weijie Wang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Ding
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weihong Ge
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yongsheng Fan
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Xu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Li Xu,
| |
Collapse
|
15
|
Effect of add-on hydroxychloroquine therapy on serum proinflammatory cytokine levels in patients with systemic lupus erythematosus. Sci Rep 2022; 12:10175. [PMID: 35715525 PMCID: PMC9205904 DOI: 10.1038/s41598-022-14571-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
We investigated the effect of hydroxychloroquine (HCQ) as an add-on treatment to immunosuppressants on the expression of proinflammatory cytokines in patients with systemic lupus erythematosus. Serum levels of tumor necrosis factor (TNF)-α, interleukin (IL)-2, IL-6, IL-8, vascular endothelial growth factor (VEGF)-A, monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1α (MIP-1α), and interleukin 1 receptor antagonist (IL-1ra) were measured immediately before and 3 months after treatment with oral HCQ. Among the 51 patients enrolled in the study, HCQ treatment led to significantly reduced serum levels of TNF-α, IL-6, IL-8, VEGF-A, IL-1ra, and IL-2 (p < 0.0001; p = 0.0006; p = 0.0460, p = 0.0177; p < 0.0001; p = 0.0282, respectively) and to decreased (but not significantly) levels of MIP-1α (p = 0.0746). No significant changes were observed in the serum MCP-1 levels before and after HCQ administration (p = 0.1402). Our results suggest that an add-on HCQ treatment modulates the expression of proinflammatory cytokines even in systemic lupus erythematosus patients with low disease activity.
Collapse
|
16
|
Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm (Beijing) 2022; 3:e147. [PMID: 35702353 PMCID: PMC9175564 DOI: 10.1002/mco2.147] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a large family of small secreted proteins that have fundamental roles in organ development, normal physiology, and immune responses upon binding to their corresponding receptors. The primary functions of chemokines are to coordinate and recruit immune cells to and from tissues and to participate in regulating interactions between immune cells. In addition to the generally recognized antimicrobial immunity, the chemokine/chemokine receptor axis also exerts a tumorigenic function in many different cancer models and is involved in the formation of immunosuppressive and protective tumor microenvironment (TME), making them potential prognostic markers for various hematologic and solid tumors. In fact, apart from its vital role in tumors, almost all inflammatory diseases involve chemokines and their receptors in one way or another. Modulating the expression of chemokines and/or their corresponding receptors on tumor cells or immune cells provides the basis for the exploitation of new drugs for clinical evaluation in the treatment of related diseases. Here, we summarize recent advances of chemokine systems in protumor and antitumor immune responses and discuss the prevailing understanding of how the chemokine system operates in inflammatory diseases. In this review, we also emphatically highlight the complexity of the chemokine system and explore its potential to guide the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| |
Collapse
|
17
|
Stern EP, Host LV, Wanjiku I, Escott KJ, Gilmour PS, Ochiel R, Unwin R, Burns A, Ong VH, Cadiou H, O'Keeffe AG, Denton CP. Zibotentan in systemic sclerosis-associated chronic kidney disease: a phase II randomised placebo-controlled trial. Arthritis Res Ther 2022; 24:130. [PMID: 35650639 PMCID: PMC9158153 DOI: 10.1186/s13075-022-02818-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/21/2022] [Indexed: 11/19/2022] Open
Abstract
Background We report results from a phase II randomised placebo-controlled trial assessing zibotentan, a highly selective endothelin receptor antagonist (ERA), in chronic kidney disease (CKD) secondary to systemic sclerosis (SSc). Methods This trial included three sub-studies: ZEBRA 1—a randomised placebo-controlled, double-blind trial of zibotentan in SSc patients with CKD2 or CKD3 (and glomerular filtration rate (GFR) >45 ml/min) over 26 weeks; ZEBRA 2A—a 26-week placebo-controlled, single-blind trial of zibotentan in scleroderma renal crisis patients not requiring dialysis; and ZEBRA 2B—an open label pharmacokinetic study of zibotentan in patients on haemodialysis. Results Sixteen patients were screened for ZEBRA 1. Of these, 6 patients were randomised to zibotentan and 7 to placebo. In ZEBRA 1, there were 47 non-serious adverse events (AE) during the trial. Twenty-seven occurred in the placebo group and 20 in the zibotentan group. One serious adverse event (SAE) occurred during ZEBRA1, in the placebo arm. Descriptive statistics did not suggest an effect of study drug on serum sVCAM1. Estimated GFR numerically declined in patients treated with placebo at 26 weeks and 52 weeks. In contrast, average eGFR increased in zibotentan-treated cases. The 4 patients in ZEBRA 2A experienced 8 non-serious AEs, distributed equally between placebo and zibotentan. There was one SAE each in placebo and zibotentan groups, both unrelated to study medication. ZEBRA 2B recruited 8 patients, 6 completed first dosing, and 2 completed a second dosing visit. Pharmacokinetic analysis confirmed zibotentan levels within the therapeutic range. Three patients experienced 3 non-serious AEs. One SAE occurred and was unrelated to study drug. Conclusions Zibotentan was generally well-tolerated. ZEBRA 1 did not show any effect of zibotentan on serum sVCAM-1 but was associated with numerical improvement in eGFR at 26 weeks that was more marked at 52 weeks. ZEBRA 2B suggested a feasible dose regimen for haemodialysis patients. Trial registration EudraCT no: 2013-003200-39 (first posted January 28, 2014) ClinicalTrials.gov Identifier: NCT02047708 Sponsor protocol number: 13/0077
Collapse
Affiliation(s)
- Edward P Stern
- Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Lauren V Host
- Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Ivy Wanjiku
- Division of Medicine, University College London, Royal Free Campus, London, UK
| | - K Jane Escott
- Emerging Innovations Unit, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Peter S Gilmour
- Emerging Innovations Unit, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Rachel Ochiel
- Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Robert Unwin
- Division of Medicine, University College London, Royal Free Campus, London, UK.,Early Clinical Development, Cardiovascular, Renal & Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Aine Burns
- Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Voon H Ong
- Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Helen Cadiou
- Joint Research Office, University College London, London, UK
| | - Aidan G O'Keeffe
- Joint Research Office, University College London, London, UK.,School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Christopher P Denton
- Division of Medicine, University College London, Royal Free Campus, London, UK. .,UCL Centre for Rheumatology and Connective Tissue Diseases, 2nd Floor - UCL Medical School Building, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
18
|
Whittall-Garcia L, Goliad K, Kim M, Bonilla D, Gladman D, Urowitz M, Fortin PR, Atenafu EG, Touma Z, Wither J. Identification and Validation of a Urinary Biomarker Panel to Accurately Diagnose and Predict Response to Therapy in Lupus Nephritis. Front Immunol 2022; 13:889931. [PMID: 35711439 PMCID: PMC9196040 DOI: 10.3389/fimmu.2022.889931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/02/2022] [Indexed: 11/15/2022] Open
Abstract
Background We have previously shown that 15 urinary biomarkers (of 129 tested by Luminex), discriminate between active Lupus Nephritis (ALN) and non-LN patients. The aim of this study was to evaluate the ability of these 15 previously-identified urinary biomarkers to predict treatment responses to conventional therapy, and for the most predictive of these biomarkers to validate their utility to identify ALN patients in an independent prospectively-acquired lupus cohort. Methods Our study had a 3-stage approach. In stage 1, we used Luminex to examine whether our previously identified urinary biomarkers at the time of the renal flare ( ± 3 months) or 12 ± 3 months after treatment of biopsy-proven ALN could predict treatment responses. In stage 2, a larger prospectively-acquired cross-sectional cohort was used to further validate the utility of the most predictive urinary biomarkers (identified in stage 1) to detect ALN patients. In this 2nd stage, cut-offs with the best operating characteristics to detect ALN patients were produced for each biomarker and different combinations and/or numbers of elevated biomarkers needed to accurately identify ALN patients were analyzed. In stage 3, we aimed to further corroborate the sensitivity of the cut-offs created in stage 2 to detect ALN patients in a biopsy-proven ALN cohort who had a urine sample collection within 3 months of their biopsy. Results Twenty-one patients were included in stage 1. Twelve (57.1%), 4 (19.1%), and 5 (23.8%) patients had a complete (CR), partial (PR) and no (NR) remission at 24 ± 3 months, respectively. The percentage decrease following 12 ± 3 months of treatment for Adiponectin, MCP-1, sVCAM-1, PF4, IL-15 and vWF was significantly higher in patients with CR in comparison to those with PR/NR. In stage 2, a total of 247 SLE patients were included, of which 24 (9.7%) had ALN, 79 (31.9%) had LN in remission (RLN) and 144 (58.3%) were non-LN (NLN) patients. Based on the combinations of biomarkers with the best operating characteristics we propose “rule out” and “rule in” ALN criteria. In stage 3, 53 biopsy-proven ALN patients were included, 35 with proliferative LN and 18 with non-proliferative ALN, demonstrating that our “rule in ALN” criteria operate better in detecting active proliferative than non-proliferative classes. Conclusions Our results provide further evidence to support the role of Adiponectin, MCP-1, sVCAM-1 and PF4 in the detection of proliferative ALN cases. We further show the clinical utility of measuring multiple rather than a single biomarker and we propose novel “rule in” and “rule out” criteria for the detection of proliferative ALN with excellent operating characteristics.
Collapse
Affiliation(s)
- Laura Whittall-Garcia
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Kirubel Goliad
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Michael Kim
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Dennisse Bonilla
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Dafna Gladman
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Murray Urowitz
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Paul R. Fortin
- Division of Rheumatology, Department of Medicine, Centre de recherche du CHU de Québec–Université Laval, Quebec City, QC, Canada
| | - Eshetu G. Atenafu
- Department of Biostatistics, University Health Network, Toronto, ON, Canada
| | - Zahi Touma
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Joan Wither
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- *Correspondence: Joan Wither,
| |
Collapse
|
19
|
Manchanda AS, Kwan AC, Ishimori M, Thomson LEJ, Li D, Berman DS, Bairey Merz CN, Jefferies C, Wei J. Coronary Microvascular Dysfunction in Patients With Systemic Lupus Erythematosus and Chest Pain. Front Cardiovasc Med 2022; 9:867155. [PMID: 35498009 PMCID: PMC9053571 DOI: 10.3389/fcvm.2022.867155] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 01/19/2023] Open
Abstract
Chest pain is a common symptom in patients with systemic lupus erythematosus, an autoimmune disease that is associated with increased cardiovascular morbidity and mortality. While chest pain mechanisms can be multifactorial and often attributed to non-coronary or non-cardiac cardiac etiologies, emerging evidence suggests that ischemia with no obstructive coronary arteries (INOCA) is a prevalent condition in patients with chest pain and no obstructive coronary artery disease. Coronary microvascular dysfunction is reported in approximately half of SLE patients with suspected INOCA. In this mini review, we highlight the cardiovascular risk assessment, mechanisms of INOCA, and diagnostic approach for patients with SLE and suspected CMD.
Collapse
Affiliation(s)
- Ashley S. Manchanda
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Alan C. Kwan
- Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Imaging, Mark Taper Imaging Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mariko Ishimori
- Division of Rheumatology and Department of Biomedical Sciences, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Louise E. J. Thomson
- Department of Imaging, Mark Taper Imaging Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Debiao Li
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Daniel S. Berman
- Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Imaging, Mark Taper Imaging Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - C. Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Caroline Jefferies
- Division of Rheumatology and Department of Biomedical Sciences, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Janet Wei
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Cedars-Sinai Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- *Correspondence: Janet Wei
| |
Collapse
|
20
|
Ngamjanyaporn P, Worawichawong S, Pisitkun P, Khiewngam K, Kantachuvesiri S, Nongnuch A, Assanatham M, Sathirapongsasuti N, Kitiyakara C. Predicting treatment response and clinicopathological findings in lupus nephritis with urine epidermal growth factor, monocyte chemoattractant protein-1 or their ratios. PLoS One 2022; 17:e0263778. [PMID: 35271583 PMCID: PMC8912200 DOI: 10.1371/journal.pone.0263778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/26/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction There is a need for sensitive and specific biomarkers to predict kidney damage and therapeutic response in lupus nephritis (LN). Monocyte chemoattractant protein-1 (MCP-1) and epidermal growth factor (EGF) are cytokines with divergent roles. EGF or EGF/MCP1 ratio have been shown to correlate with prognosis in primary glomerulonephritis, but there is limited information in lupus nephritis (LN). This study evaluated the roles of MCP-1, EGF or their ratio as biomarkers of histopathology and response to treatment in LN. Methods This was a cross-sectional and observational study. Baseline urine MCP-1 and EGF levels in systemic lupus erythematosus (SLE) patients and controls (total n = 101) were compared, and levels were correlated with clinicopathological findings and subsequent response to treatment. Results MCP-1 was higher in active LN (n = 69) compared to other SLE groups and controls, whereas EGF was not different. MCP-1 correlated with disease activity (proteinuria, renal SLEDAI, classes III/IV/V, and high activity index.) By contrast, EGF correlated with eGFR, but not with proteinuria, activity index, or class III/IV/V. MCP-1 was higher, and EGF was lower in high chronicity index. EGF/MCP-1 decreased with greater clinicopathological severity. In a subgroup with proliferative LN who completed six months of induction therapy (n = 41), EGF at baseline was lower in non-responders compared to responders, whereas MCP-1 was similar. By multivariable analysis, baseline EGF was independently associated with subsequent treatment response. Area under the curve for EGF to predict response was 0.80 (0.66–0.95). EGF ≥ 65.6 ng/ mgCr demonstrated 85% sensitivity and 71% specificity for response. EGF/MCP-1 did not improve the prediction for response compared to EGF alone. Conclusion MCP-1 increased with disease activity, whereas EGF decreased with low GFR and chronic damage. Urine EGF may be a promising biomarker to predict therapeutic response in LN. EGF/MCP-1 did not improve the prediction of response.
Collapse
Affiliation(s)
- Pintip Ngamjanyaporn
- Faculty of Medicine Ramathibodi Hospital, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Suchin Worawichawong
- Faculty of Medicine Ramathibodi Hospital, Department of Pathology, Mahidol University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Faculty of Medicine Ramathibodi Hospital, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Khantong Khiewngam
- Faculty of Medicine Ramathibodi Hospital, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Surasak Kantachuvesiri
- Faculty of Medicine Ramathibodi Hospital, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Arkom Nongnuch
- Faculty of Medicine Ramathibodi Hospital, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Montira Assanatham
- Faculty of Medicine Ramathibodi Hospital, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Nuankanya Sathirapongsasuti
- Faculty of Medicine Ramathibodi Hospital, Section of Translational Medicine, Mahidol University, Bangkok, Thailand
| | - Chagriya Kitiyakara
- Faculty of Medicine Ramathibodi Hospital, Department of Medicine, Mahidol University, Bangkok, Thailand
- * E-mail: ,
| |
Collapse
|
21
|
Tejon G, Valdivieso N, Flores-Santibañez F, Barra-Valdebenito V, Martínez V, Rosemblatt M, Sauma D, Bono MR. Phenotypic and functional alterations of peritoneal macrophages in lupus-prone mice. Mol Biol Rep 2022; 49:4193-4204. [PMID: 35211864 PMCID: PMC9262788 DOI: 10.1007/s11033-022-07252-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/09/2022] [Indexed: 11/28/2022]
Abstract
Background Several studies have demonstrated the contribution of innate immune cells, including macrophages, in promoting systemic lupus erythematosus (SLE). Macrophages, one of the most abundant cell populations in the peritoneal cavity, are considered multifunctional cells with phenotypic plasticity. However, the functional properties of peritoneal macrophages in steady-state and during the progression of SLE remain poorly defined. Methods and results Using the [NZB × NZW]F1 (BWF1) murine model of SLE, we analyzed the phenotype and function of peritoneal macrophages during the disease’s onset. We found a higher frequency of peritoneal macrophages and B1a cells in BWF1-diseased mice than age-matched controls. Additionally, macrophages from diseased animals expressed lower levels of CD206, MHC-II, and Sirpα. RNAseq analysis identified 286 differentially expressed genes in peritoneal macrophages from diseased-BWF1 mice compared to control mice. Functional experiments demonstrate that peritoneal macrophages from diseased-BWF1 mice secrete higher levels of pro-inflammatory cytokines when activated with TLR7 and TLR9 agonists, and they were less efficient in suppressing the activation and proliferation of peritoneal LPS-activated B cells. These data demonstrate that peritoneal macrophages from BWF1-diseased mice present phenotypic and functional alterations shifting to a more pro-inflammatory state. Conclusions The increase of macrophages with an altered phenotype and function together with the accumulation of B1a cells in the peritoneal cavity of diseased-BWF1 mice may promote the progression of the disease. Advancing awareness of the role and phenotype of peritoneal macrophages in SLE may contribute to a better understanding of these types of diseases and the development of novel therapies. Supplementary Information The online version contains supplementary material available at 10.1007/s11033-022-07252-0.
Collapse
Affiliation(s)
- Gabriela Tejon
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Nicolás Valdivieso
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | | | | | - Víctor Martínez
- FAVET-INBIOGEN, Facultad de Ciencias Veterinarias, Universidad de Chile, Santiago, Chile
| | - Mario Rosemblatt
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - María Rosa Bono
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.
- Centro Ciencia & Vida, Santiago, Chile.
| |
Collapse
|
22
|
Valiente GR, Munir A, Hart ML, Blough P, Wada TT, Dalan EE, Willis WL, Wu LC, Freud AG, Jarjour WN. Gut dysbiosis is associated with acceleration of lupus nephritis. Sci Rep 2022; 12:152. [PMID: 34996983 PMCID: PMC8742035 DOI: 10.1038/s41598-021-03886-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/01/2021] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota (GM) exerts a strong influence over the host immune system and dysbiosis of this microbial community can affect the clinical phenotype in chronic inflammatory conditions. To explore the role of the GM in lupus nephritis, we colonized NZM2410 mice with Segmented Filamentous Bacteria (SFB). Gut colonization with SFB was associated with worsening glomerulonephritis, glomerular and tubular immune complex deposition and interstitial inflammation compared to NZM2410 mice free of SFB. With SFB colonization mice experienced an increase in small intestinal lamina propria Th17 cells and group 3 innate lymphoid cells (ILC3s). However, although serum IL-17A expression was elevated in these mice, Th17 cells and ILC3s were not detected in the inflammatory infiltrate in the kidney. In contrast, serum and kidney tissue expression of the macrophage chemoattractants MCP-1 and CXCL1 were significantly elevated in SFB colonized mice. Furthermore, kidney infiltrating F4/80+CD206+M2-like macrophages were significantly increased in these mice. Evidence of increased gut permeability or "leakiness" was also detected in SFB colonized mice. Finally, the intestinal microbiome of SFB colonized mice at 15 and 30 weeks of age exhibited dysbiosis when compared to uncolonized mice at the same time points. Both microbial relative abundance as well as biodiversity of colonized mice was found to be altered. Collectively, SFB gut colonization in the NZM2410 mouse exacerbates kidney disease, promotes kidney M2-like macrophage infiltration and overall intestinal microbiota dysbiosis.
Collapse
Affiliation(s)
- Giancarlo R Valiente
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA
| | - Armin Munir
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Perry Blough
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Takuma T Wada
- Saitama Medical University, Moroyama, Saitama, Japan
| | - Emma E Dalan
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - William L Willis
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lai-Chu Wu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, USA
| | - Aharon G Freud
- The Department of Pathology and the James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Wael N Jarjour
- Department of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
23
|
OUP accepted manuscript. J Appl Lab Med 2022; 7:1450-1467. [DOI: 10.1093/jalm/jfac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 04/25/2022] [Indexed: 11/14/2022]
|
24
|
Singh S, Anshita D, Ravichandiran V. MCP-1: Function, regulation, and involvement in disease. Int Immunopharmacol 2021; 101:107598. [PMID: 34233864 PMCID: PMC8135227 DOI: 10.1016/j.intimp.2021.107598] [Citation(s) in RCA: 444] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/25/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023]
Abstract
MCP-1 (Monocyte chemoattractant protein-1), also known as Chemokine (CC-motif) ligand 2 (CCL2), is from family of CC chemokines. It has a vital role in the process of inflammation, where it attracts or enhances the expression of other inflammatory factors/cells. It leads to the advancement of many disorders by this main mechanism of migration and infiltration of inflammatory cells like monocytes/macrophages and other cytokines at the site of inflammation. MCP-1 has been inculpated in the pathogenesis of numerous disease conditions either directly or indirectly like novel corona virus, cancers, neuroinflammatory diseases, rheumatoid arthritis, cardiovascular diseases. The elevated MCP-1 level has been observed in COVID-19 patients and proven to be a biomarker associated with the extremity of disease along with IP-10. This review will focus on involvement and role of MCP-1 in various pathological conditions.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India.
| | - D Anshita
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP), Zandaha Road, Hajipur, Bihar, India
| |
Collapse
|
25
|
Wang ZH, Dai ZW, Dong YY, Wang H, Yuan FF, Wang B, Ye DQ. Urinary Tumor Necrosis Factor-Like Weak Inducer of Apoptosis as a Biomarker for Diagnosis and Evaluating Activity in Lupus Nephritis: A Meta-analysis. J Clin Rheumatol 2021; 27:272-277. [PMID: 32028305 DOI: 10.1097/rhu.0000000000001316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Urinary tumor necrosis factor-like weak inducer of apoptosis (uTWEAK) has been identified as a candidate biomarker for lupus nephritis (LN). However, its diagnostic value remains unclear. This meta-analysis was conducted to comprehensively evaluate the value of uTWEAK for diagnosis and evaluating activity in LN. METHODS Medline, Web of Science, Chinese Biomedical Medical, and Chinese National Knowledge Infrastructure databases were searched to acquire eligible studies published before September 30, 2019. The quality of the studies was evaluated by Quality Assessment of Diagnostic Accuracy Studies-2. Summary receiver operating characteristic curve and area under the curve were applied to summarize the overall diagnostic performances. The pooled sensitivity, specificity, and diagnostic odds ratio (DOR) were calculated with the fixed-effects model. RevMan 5.3, Stata 12.0, and Meta-disc 1.4 software were used. RESULTS A total of 7 studies were included. Of these, 4 studies were available for comparison between SLE with and without LN, and 3 studies were for active and inactive LN. The total area under the curve was 0.8640, and DOR was 14.89 (95% confidence interval [CI], 7.95-27.86). For LN diagnosis, the pooled sensitivity, specificity, and DOR were 0.55 (95% CI, 0.47-0.63), 0.92 (95% CI, 0.86-0.96), and 16.54 (95% CI, 7.57-36.15), respectively. For assessing LN activity, the pooled sensitivity, specificity, and DOR were 0.91 (95% CI, 0.82-0.96), 0.70 (95% CI, 0.58-0.81), and 18.45 (95% CI, 7.45-45.87), respectively. CONCLUSIONS This meta-analysis indicated that uTWEAK has relatively moderate sensitivity and specificity for diagnosis and evaluating activity in LN, suggesting that uTWEAK can serve as a helpful biomarker for LN.
Collapse
|
26
|
Immune-Related Urine Biomarkers for the Diagnosis of Lupus Nephritis. Int J Mol Sci 2021; 22:ijms22137143. [PMID: 34281193 PMCID: PMC8267641 DOI: 10.3390/ijms22137143] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/17/2022] Open
Abstract
The kidney is one of the main organs affected by the autoimmune disease systemic lupus erythematosus. Lupus nephritis (LN) concerns 30-60% of adult SLE patients and it is significantly associated with an increase in the morbidity and mortality. The definitive diagnosis of LN can only be achieved by histological analysis of renal biopsies, but the invasiveness of this technique is an obstacle for early diagnosis of renal involvement and a proper follow-up of LN patients under treatment. The use of urine for the discovery of non-invasive biomarkers for renal disease in SLE patients is an attractive alternative to repeated renal biopsies, as several studies have described surrogate urinary cells or analytes reflecting the inflammatory state of the kidney, and/or the severity of the disease. Herein, we review the main findings in the field of urine immune-related biomarkers for LN patients, and discuss their prognostic and diagnostic value. This manuscript is focused on the complement system, antibodies and autoantibodies, chemokines, cytokines, and leukocytes, as they are the main effectors of LN pathogenesis.
Collapse
|
27
|
Kot K, Kupnicka P, Witulska O, Czepan A, Łanocha-Arendarczyk NA, Łanocha AA, Kosik-Bogacka DI. Potential Biomarkers in Diagnosis of Renal Acanthamoebiasis. Int J Mol Sci 2021; 22:6583. [PMID: 34205319 PMCID: PMC8234237 DOI: 10.3390/ijms22126583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 12/05/2022] Open
Abstract
Recent studies indicate that Acanthamoeba spp. may play a significant role in kidney dysfunction. The aim of the study was to examine the levels of kidney injury molecule 1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and monocyte chemotactic protein 1 (MCP-1), as well as an activity of matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9, respectively) in the kidneys of immunocompetent and immunosuppressed mice infected with Acanthamoeba spp. The levels of KIM-1, NGAL, and MCP-1 were analyzed by enzyme-linked immunosorbent assay (ELISA), and the activity of MMPs was determined by gelatin zymography. The elevated KIM-1 level was found in the kidneys of immunocompetent mice at the beginning of Acanthamoeba spp. infection. In the immunosuppressed mice, the KIM-1 level was statistically different. The statistically decreased NGAL level was found in the kidneys of immunocompetent mice compared to the uninfected mice. In the immunocompromised mice, we found statistically significant differences in MCP-1 levels between the uninfected and infected groups. There was an increase in the expression of both MMP-2 and MMP-9 in the kidneys of immunocompetent and immunosuppressed mice infected with Acanthamoeba spp. compared to the uninfected mice. The results indicate that KIM-1, NGAL, MCP-1, MMP-2, MMP-9, and MMP-9/NGAL might be promising biomarkers of renal acanthamoebiasis.
Collapse
Affiliation(s)
- Karolina Kot
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (K.K.); (O.W.); (A.C.); (N.A.Ł.-A.)
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Oliwia Witulska
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (K.K.); (O.W.); (A.C.); (N.A.Ł.-A.)
| | - Aleksandra Czepan
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (K.K.); (O.W.); (A.C.); (N.A.Ł.-A.)
| | - Natalia Agnieszka Łanocha-Arendarczyk
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (K.K.); (O.W.); (A.C.); (N.A.Ł.-A.)
| | - Aleksandra Anna Łanocha
- Department of Haematology and Transplantology, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Danuta Izabela Kosik-Bogacka
- Independent Laboratory of Pharmaceutical Botany, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| |
Collapse
|
28
|
Capecchi R, Puxeddu I, Pratesi F, Migliorini P. New biomarkers in SLE: from bench to bedside. Rheumatology (Oxford) 2021; 59:v12-v18. [PMID: 32911542 PMCID: PMC7719038 DOI: 10.1093/rheumatology/keaa484] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/23/2020] [Indexed: 12/20/2022] Open
Abstract
Biomarkers may have a diagnostic or monitoring value, or may predict response to therapy or disease course. The aim of this review is to discuss new serum and urinary biomarkers recently proposed for the diagnosis and management of SLE patients. Novel sensitive and specific assays have been proposed to evaluate complement proteins, ‘old’ biomarkers that are still a cornerstone in the management of this disease. Chemokines and lectins have been evaluated as surrogate biomarkers of IFN signature. Other cytokines like the B cell activating factor (BAFF) family cytokines are directly related to perturbations of the B cell compartment as key pathogenetic mechanism of the disease. A large number of urine biomarkers have been proposed, either related to the migration and homing of leukocytes to the kidney or to the local regulation of inflammatory circuits and the survival of renal intrinsic cells. The combination of traditional disease-specific biomarkers and novel serum or urine biomarkers may represent the best choice to correctly classify, stage and treat patients with SLE.
Collapse
Affiliation(s)
- Riccardo Capecchi
- Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ilaria Puxeddu
- Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Federico Pratesi
- Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Paola Migliorini
- Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
29
|
Moloi MW, Rusch JA, Omar F, Ekrikpo U, Dandara C, Bello AK, Jayne D, Okpechi IG. Urinary MCP-1 and TWEAK as non-invasive markers of disease activity and treatment response in patients with lupus nephritis in South Africa. Int Urol Nephrol 2021; 53:1865-1873. [PMID: 33459955 DOI: 10.1007/s11255-020-02780-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/26/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Treatment of patients with lupus nephritis (LN) requires judicious use of immunosuppression. Novel biomarkers may be useful for monitoring disease activity and treatment response. We assessed the utility of urinary monocyte chemoattractant protein-1 (uMCP-1) and urinary tumour necrosis factor-like weak inducer of apoptosis (uTWEAK) for disease activity and treatment response monitoring in South Africans with LN. METHODS We recruited consenting patients with active LN confirmed on kidney biopsy. Urinary levels of MCP-1 and TWEAK were assayed at baseline and after completion of induction therapy using ELISA methods. We also collected relevant demographic, clinical and biochemical data for patients included in this study. RESULTS The mean age of patients in this study was 29.8 ± 10.7 years, 60% were patients of mixed ancestry, 70% had proliferative LN and mean spot urine proteinuria at baseline was 0.37 (0.18-0.59) g/mmolCr. At completion of induction therapy, the level of uMCP-1 had reduced to 314.5 (IQR: 197.0-622) pg/mgCr from a baseline of 1092.7 (IQR 578.6-1848) pg/mgCr (P = 0.06) while uTWEAK had reduced to 36.0 (IQR 17.0-88.0) pg/mgCr from 159.0 (IQR: 88.5-295.5) pg/mgCr (P = 0.03). For patients reaching early complete or partial remission (n = 17), both biomarkers had significantly declined in their urine: uMCP-1 (P = 0.018) and uTWEAK (P = 0.015). There was no reduction of both biomarkers in patients not achieving remission and no association between uMCP-1 or uTWEAK with renal histological features. CONCLUSION Our study shows that uMCP-1 and uTWEAK are elevated in patients with active LN, correlated with the remission status (response to treatment) at the end of induction therapy and can, therefore, be useful for monitoring disease activity and treatment response.
Collapse
Affiliation(s)
- Mothusi W Moloi
- Division of Nephrology and Hypertension, University of Cape Town, Cape Town, South Africa.,Kidney and Hypertension Research Unit, University of Cape Town Cape Town, Cape Town, South Africa.,Department of Medicine, University of Botswana, Gaborone, Botswana
| | - Jody A Rusch
- Division of Chemical Pathology, University of Cape Town and National Health Laboratory Services (NHLS), Cape Town, South Africa
| | - Fierdoz Omar
- Division of Chemical Pathology, University of Cape Town and National Health Laboratory Services (NHLS), Cape Town, South Africa
| | - Udeme Ekrikpo
- Division of Nephrology and Hypertension, University of Cape Town, Cape Town, South Africa.,Kidney and Hypertension Research Unit, University of Cape Town Cape Town, Cape Town, South Africa.,Department of Medicine, University of Uyo, Uyo, Nigeria
| | - Collet Dandara
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Aminu K Bello
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ikechi G Okpechi
- Division of Nephrology and Hypertension, University of Cape Town, Cape Town, South Africa. .,Kidney and Hypertension Research Unit, University of Cape Town Cape Town, Cape Town, South Africa. .,Department of Medicine, University of Alberta, Edmonton, Canada.
| |
Collapse
|
30
|
Liu L, Wang R, Ding H, Tian L, Gao T, Bao C. The utility of urinary biomarker panel in predicting renal pathology and treatment response in Chinese lupus nephritis patients. PLoS One 2020; 15:e0240942. [PMID: 33108403 PMCID: PMC7591050 DOI: 10.1371/journal.pone.0240942] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023] Open
Abstract
Given the urgent need for non-invasive biomarkers of LN, we aim to identify novel urinary biomarkers that facilitate diagnosis, assessment of disease activity and prediction of treatment response in a retrospective SLE cohort. A total of 154 SLE patients and 55 healthy controls were enrolled, among whom 73 were active LN patients. We measured renal activity by renal SLEDAI. The treatment response of the active LN patients who finished 6-month induction therapy was assessed based on the American College of Rheumatology response criteria. The expression levels of 10 urinary biomarkers (UBMs): β2-MG, calbindin D, cystatin C, IL-18, KIM-1, MCP-1, nephrin, NGAL, VCAM-1, and VDBP were tested using Luminex high-throughput proteomics technology. All but urinary nephrin levels were significantly increased in active LN compared to healthy controls. uCystatinC, uMCP-1, uKIM-1 levels were significantly higher in active LN group compared to inactive LN group. Correlation analysis revealed positive correlation between uCystatinC, uKIM-1, uMCP-1, uNGAL, uVDBP and RSLEDAI score. In renal pathology, uCystatinC, uKIM-1, uVCAM-1, and uVDBP positively correlated with activity index (AI) while uVCAM-1 positively correlated with chronicity index (CI). Moreover, the combination of uVCAM-1, uCystatinC, uKIM-1 discriminated proliferative LN from membranous LN with an AUC of 0.80 (95%CI: 0.69–0.90). Most importantly, baseline uNGAL demonstrated good prediction ability to discriminate responders from non-responders in active LN patients after 6-month induction therapy. Using a multiplex bead technique, we have identified the combination of uVCAM-1, uCystatinC, uKIM-1 as a biomarker panel to reflect renal pathology and NGAL as a promising urinary biomarker to both reflect disease activity and predict treatment response.
Collapse
Affiliation(s)
- Li Liu
- Department of Emergency Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Wang
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huihua Ding
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Tian
- Department of Emergency Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Gao
- Department of Emergency Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunde Bao
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
31
|
Brady MP, Korte EA, Caster DJ, Powell DW. TNIP1/ABIN1 and lupus nephritis: review. Lupus Sci Med 2020; 7:e000437. [PMID: 33122334 PMCID: PMC7597513 DOI: 10.1136/lupus-2020-000437] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/15/2020] [Accepted: 10/08/2020] [Indexed: 12/17/2022]
Abstract
SLE is a complex autoimmune disease with genetic, epigenetic, immune-regulatory, environmental and hormonal factors. Kidney inflammation and injury, termed lupus nephritis (LN), occurs in over half of patients with SLE and is a leading cause of disability and death. There is a high degree of short-term and long-term side effects associated with current LN therapies and they are not effective for many patients. Thus, novel therapies with reduced toxicity and improved efficacy are drastically needed. Many of the known LN susceptibility genes have functions that mediate inflammation via cytokine/chemokine production and activation of myeloid and B cells. Understanding the cellular and molecular mechanisms mediated by these variant gene products provides valuable insight for the development of improved and personalised diagnostics and therapeutics. This review describes variants in the TNIP1 (tumour necrosis factor α-induced protein 3-interacting protein 1) gene associated with risks for SLE and LN and potential roles for loss of function of its protein product ABIN1 in the activation of myeloid and B-cell-mediated injury in LN.
Collapse
Affiliation(s)
- Makayla P Brady
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Erik A Korte
- Bluewater Diagnostics Laboratory, Mt. Washington, Kentucky, USA
| | - Dawn J Caster
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - David W Powell
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
32
|
Clinical Use of Complement, Inflammation, and Fibrosis Biomarkers in Autoimmune Glomerulonephritis. Kidney Int Rep 2020; 5:1690-1699. [PMID: 33102961 PMCID: PMC7569694 DOI: 10.1016/j.ekir.2020.07.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/04/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Complement activation, inflammation, and fibrosis play central roles in the mechanisms of injury in autoimmune glomerulonephritis (GN) but they are seldom assessed in epidemiologic studies. The measurement of urinary biomarkers of these pathways of injury could parallel disease activity and add clinical value beyond proteinuria. Methods We performed a prospective cohort study of 100 patients with focal and segmental glomerulosclerosis (FSGS), membranous nephropathy (MN), IgA nephropathy (IgAN), lupus nephritis (LN), anti-neutrophil cytoplasmic autoantibody-associated vasculitis (AAV), and membranoproliferative GN (MPGN) followed for 33 (18-54) months. Repeated urinary samples were collected throughout their follow-up to determine proteinuria, urinary sC5b-9, monocyte chemoattractant protein-1 (MCP-1), and transforming growth factor-beta 1 (TGF-β1), expressed as creatinine ratios. We identified 177 periods of active and inactive disease based on current remission definitions for each disease. Results Urinary sC5b-9, MCP-1, and TGF-β1 were present in each disease. In periods leading to a remission, the reduction of urinary sC5b-9 was 91%, greater than for proteinuria with 76%. During inactive periods, those who did not experience a relapse maintained lower levels of biomarkers compared with those who relapsed. At that time, the increase in urinary sC5b-9 was significantly greater than the rise in proteinuria (8.5-fold increase compared with 3.2-fold) and urinary MCP-1 and TGF-β1. Using current remission definitions for each disease, thresholds for each biomarker were determined using receiver operating characteristic curves. Individuals who averaged levels below these cutoffs during their follow-up had better renal outcomes. Conclusion In autoimmune glomerular diseases, urinary sC5b-9, MCP-1, and TGF-β1 are present and parallel disease activity and outcomes. Urinary sC5b-9 appears to be a more discerning marker of immunologic remissions and relapses.
Collapse
|
33
|
Erez DL, Denburg MR, Afolayan S, Jodele S, Wallace G, Davies SM, Seif AE, Bunin N, Laskin BL, Sullivan KE. Acute Kidney Injury in Children after Hematopoietic Cell Transplantation Is Associated with Elevated Urine CXCL10 and CXCL9. Biol Blood Marrow Transplant 2020; 26:1266-1272. [PMID: 32165324 DOI: 10.1016/j.bbmt.2020.02.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is nearly universally associated with worse outcomes, especially among children after hematopoietic stem cell transplant (HCT). Our objective was to examine urinary immune biomarkers of AKI after HCT to provide insights into novel mechanisms of kidney injury in this population. Studying patients undergoing allogeneic HCT provides a unique opportunity to examine immune markers of AKI because the risk of AKI is high and the immune system newly develops after transplant. Children (>2 years old) and young adults undergoing their first allogeneic HCT and enrolled in a prospective, observational cohort study at 2 large children's hospitals had urine collected pre-HCT and monthly for the first 4 months after HCT. Urine samples at each monthly time point were assayed for 8 immune-related biomarkers. AKI was defined as a 1.5-fold increase in the monthly serum creatinine value, which was recorded ±1 day from when the research urine sample was obtained, as compared with the pre-HCT baseline. Generalized estimating equation regression analysis evaluated the association between the monthly repeated measures (urinary biomarkers and AKI). A total of 176 patients were included from 2 pediatric centers. Thirty-six patients from 1 center were analyzed as a discovery cohort and the remaining 140 patients from the second center were analyzed as a validation cohort. AKI rates were 18% to 35% depending on the monthly time point after HCT. Urine CXCL10 and CXCL9 concentrations were significantly higher among children who developed AKI compared with children who did not (P < .01) in both cohorts. In order to gain a better understanding of the cellular source for these biomarkers in the urine, we also analyzed in vitro expression of CXCL10 and CXCL9 in kidney cell lines after stimulation with interferon-γ and interferon-α. HEK293-epithelial kidney cells demonstrated interferon-induced expression of CXCL10 and CXCL9, suggesting a potential mechanism driving the key finding. CXCL10 and CXCL9 are associated with AKI after HCT and are therefore promising biomarkers to guide improved diagnostic and treatment strategies for AKI in this high-risk population.
Collapse
Affiliation(s)
- Daniella Levy Erez
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Michelle R Denburg
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Simisola Afolayan
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Gregory Wallace
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alix E Seif
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nancy Bunin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Benjamin L Laskin
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kathleen E Sullivan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Bona N, Pezzarini E, Balbi B, Daniele SM, Rossi MF, Monje AL, Basiglio CL, Pelusa HF, Arriaga SMM. Oxidative stress, inflammation and disease activity biomarkers in lupus nephropathy. Lupus 2020; 29:311-323. [PMID: 32063098 DOI: 10.1177/0961203320904784] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lupus nephropathy is a severe and frequent complication of systemic lupus erythematosus. Here, we assessed the biomarkers of oxidative stress, inflammation and disease activity in patients with lupus nephritis. Thirty-four patients with active lupus nephritis, 31 patients with inactive lupus nephritis and 20 lupus patients without renal damage (non-lupus nephritis) were studied. Oxidative stress biomarkers malonyldialdehyde, oxidized-to-total glutathione, catalase, superoxide dismutase and total antioxidant status were assessed, as well as inflammation biomarkers CRP, interleukin 6 and monocyte chemoattractant protein 1. Renal tubular disease biomarkers neutrophil gelatinase-associated lipocalin and β2-microglobulin were assessed, together with the classic disease activity biomarkers urinary protein/creatinine ratio, anti-dsDNA, anti-C1q antibody and complement proteins C3 and C4. Significant differences were found between active lupus nephritis and inactive lupus nephritis patients and between active lupus nephritis and non-lupus nephritis patients for all the assessed biomarkers (P < 0.05), except for catalase, superoxide dismutase and interleukin 6. There is an imbalance in the redox status in active lupus nephritis patients that would be involved in lipid peroxidation of the glomerular basal membrane that would alter its integrity and could also affect renal tubular function in these patients.
Collapse
Affiliation(s)
- N Bona
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina
| | - E Pezzarini
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina
| | - B Balbi
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina
| | - S M Daniele
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina
| | - M F Rossi
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina
| | - A L Monje
- Área Clínica Médica, Facultad de Ciencias Médicas, Rosario, Argentina.,Carrera Universitaria de Nefrología, Facultad de Ciencias Médicas, Rosario, Argentina
| | - C L Basiglio
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina.,Instituto de Fisiología Experimental (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina
| | - H F Pelusa
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina
| | - S M M Arriaga
- Area Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Rosario, Argentina.,Consejo de Investigaciones de la Universidad Nacional de Rosario (CIUNR). Universidad Nacional de Rosario. Rosario, Argentina
| |
Collapse
|
35
|
Cai X, Zhu Q, Wu T, Zhu B, Liu S, Liu S, Aierken X, Ahmat A, Li N. Association of circulating resistin and adiponectin levels with Kawasaki disease: A meta-analysis. Exp Ther Med 2019; 19:1033-1041. [PMID: 32010266 PMCID: PMC6966156 DOI: 10.3892/etm.2019.8306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
The present meta-analysis was performed to examine the association between circulating blood adipokine levels and Kawasaki disease (KD). Studies were identified by searching various databases, including Web of Science, EMBASE, PubMed, Wanfang and China National Knowledge Infrastructure. After the studies were pooled, the mean difference (MD) and corresponding 95% CI were calculated. Subgroup analyses and publication bias detection were also performed. The Cochrane Q test and I2 statistics were performed using Review Manager software (version 5.3) to test for heterogeneity. A Begg's test was used to assess publication bias and STATA software (version 12.0) was used for statistical analysis. The results revealed that the KD group exhibited higher levels of resistin compared with those in the healthy controls or disease controls (non-KD; MD=20.76, 95% CI=16.16-25.36, P<0.001; MD=21.27, 95% CI=14.24-28.29, P<0.001, respectively). In addition, when compared with those in patients exhibiting non-coronary artery lesions (NCAL), those with coronary artery lesions (CAL) had higher levels of adiponectin and resistin (MD=1.00, 95% CI=0.06-1.96, P=0.04; MD=2.77, 95% CI=1.32-4.22, P<0.001). Furthermore, compared with those in the inactive-phase group, patients in the active-phase group exhibited higher levels of resistin (MD=17.73, 95% CI=12.82-22.65, P<0.001). In conclusion, the present meta-analysis indicated that resistin levels were elevated in patients with KD. It was also revealed that circulating resistin and adiponectin levels in the CAL group were significantly increased compared with those in patients with NCAL. Furthermore, the active group had higher levels of resistin than the inactive group. The results of these meta-analyses indicated that resistin may serve an important role in the pathogenesis of KD and may therefore be used as biomarkers for the diagnosis of KD, whereas adiponectin may only serve an important role in the pathogenesis of CAL and may therefore be used as a biomarker to distinguish CAL from NCAL.
Collapse
Affiliation(s)
- Xintian Cai
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Qing Zhu
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Ting Wu
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Bin Zhu
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Shasha Liu
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Shanshan Liu
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Xiayire Aierken
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Ayguzal Ahmat
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| | - Nanfang Li
- Center for Hypertension of The People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang, Urumqi, Xinjiang 830001, P.R. China
| |
Collapse
|
36
|
Tajima S, Yamamoto N, Masuda S. Clinical prospects of biomarkers for the early detection and/or prediction of organ injury associated with pharmacotherapy. Biochem Pharmacol 2019; 170:113664. [PMID: 31606409 DOI: 10.1016/j.bcp.2019.113664] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/08/2019] [Indexed: 12/31/2022]
Abstract
Several biomarkers are used to monitor organ damage caused by drug toxicity. Traditional markers of kidney function, such as serum creatinine and blood urea nitrogen are commonly used to estimate glomerular filtration rate. However, these markers have several limitations including poor specificity and sensitivity. A number of serum and urine biomarkers have recently been described to detect kidney damage caused by drugs such as cisplatin, gentamicin, vancomycin, and tacrolimus. Neutrophil gelatinase-associated lipocalin (NGAL), liver-type fatty acid-binding protein (L-FABP), kidney injury molecule-1 (KIM-1), monocyte chemotactic protein-1 (MCP-1), and cystatin C have been identified as biomarkers for early kidney damage. Hy's Law is widely used as to predict a high risk of severe drug-induced liver injury caused by drugs such as acetaminophen. Recent reports have indicated that glutamate dehydrogenase (GLDH), high-mobility group box 1 (HMGB-1), Keratin-18 (k18), MicroRNA-122 and ornithine carbamoyltransferase (OCT) are more sensitive markers of hepatotoxicity compared to the traditional markers including the blood levels of amiotransferases and total bilirubin. Additionally, the rapid development of proteomic technologies in biofluids and tissue provides a new multi-marker panel, leading to the discovery of more sensitive biomarkers. In this review, an update topics of biomarkers for the detection of kidney or liver injury associated with pharmacotherapy.
Collapse
Affiliation(s)
- Soichiro Tajima
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Nanae Yamamoto
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Satohiro Masuda
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan; Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Pharmacy, International University of Health and Welfare Narita Hospital, Japan; Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, International University of Health and Welfare Narita Hospital, Japan.
| |
Collapse
|
37
|
Caster DJ, Powell DW. Utilization of Biomarkers in Lupus Nephritis. Adv Chronic Kidney Dis 2019; 26:351-359. [PMID: 31733719 DOI: 10.1053/j.ackd.2019.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 08/22/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
Lupus nephritis (LN) occurs in up to 60% of SLE patients, and is a leading cause of disability and death. Current treatment of LN consists of a combination of high dose corticosteroids that non-specifically decrease inflammation and cytotoxic medications that reduce auto-antibody production. That combination of therapy is associated with significant side effects while remission rates remain inadequate. Since the introduction of biologics into the pharmacological armamentarium, there has been hope for less toxic and more effective therapies for LN. Unfortunately, after multiple clinical trials, no biologic has improved efficacy over standard of care therapies for LN. This is likely, in part, due to disease heterogeneity. The utilization of biomarkers in LN may provide a way to stratify patients and guide therapeutic options. In this review, we summarize traditional and novel LN biomarkers and discuss how they may be used to diagnose, stratify, and guide therapy in patients with LN, bringing precision medicine to the forefront of LN therapy.
Collapse
|
38
|
Innate Immune Dysregulation in the Development of Cardiovascular Disease in Lupus. Curr Rheumatol Rep 2019; 21:46. [DOI: 10.1007/s11926-019-0842-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Abstract
Is systemic lupus erythematosus (SLE) is occurring more frequently now than in decades past? Despite improvements in the identification of patients with SLE, the development of new classification criteria, and the recognition of several biomarkers used alone or in combination, the diagnosis of SLE is still a challenge for clinicians, in particular early in the course of the disease, which makes the recognition of secular trends difficult to ascertain. Lacking a uniform definition of preclinical lupus or incomplete lupus, it is difficult to predict accurately which patients would go on to develop SLE. We will briefly review the classification criteria, early or preclinical SLE, the epidemiology of SLE, antinuclear antibodies-negative SLE, and biomarkers of the disease.
Collapse
Affiliation(s)
- M F Ugarte-Gil
- Rheumatology Department, Hospital Guillermo Almenara Irigoyen, EsSalud, Lima, Perú
- School of Medicine, Universidad Científica del Sur, Lima, Perú
| | - L A González
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - G S Alarcón
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, USA
- School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
40
|
Adhya Z, El Anbari M, Anwar S, Mortimer A, Marr N, Karim MY. Soluble TNF-R1, VEGF and other cytokines as markers of disease activity in systemic lupus erythematosus and lupus nephritis. Lupus 2019; 28:713-721. [DOI: 10.1177/0961203319845487] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Current non-invasive methods of assessing disease activity in systemic lupus erythematosus (SLE) are of limited sensitivity and specificity. Testing includes acute phase markers, autoantibodies and complement levels. Although measurements of dsDNA antibodies and complement C3/C4 levels are routine, they remain of limited value. Improved blood and urine markers may help in early detection of flare, distinction between flare and chronic damage, and monitoring response to therapy. Methods A total of 87 patients with SLE were tested for the following cytokines in serum and urine: monocyte chemoattractant protein 1 (MCP-1), regulated upon activation, normal T cell expressed and secreted (RANTES), soluble tumour necrosis factor receptor 1 (sTNF-R1), interferon-inducible protein 10 (IP-10), monocyte inhibitory protein 1α (MIP-1α) and vascular endothelial growth factor (VEGF). Patients attending the Lupus Unit at St Thomas’ Hospital, London, UK were divided into active lupus nephritis (LN), inactive LN and non-renal SLE groups based on their renal pathology and SLE disease activity index (SLEDAI). Cytokine testing was performed using the FIDIS multiplex bead assay. Results The mean level of serum sTNF-R1 was higher in the active LN group compared with both inactive LN and non-renal SLE groups ( p < 0.001). For urine measurements there were significant differences between active LN and non-renal SLE for VEGF ( p = 0.016), after statistical correction for multiple testing. Both urinary and serum sTNF-R1 and IP-10 levels correlated with SLEDAI scores ( p < 0.001), while serum VEGF correlated weakly with SLEDAI ( p = 0.025). The optimum combination for differentiating active from inactive LN patients was serum VEGF, sTNF-R1, MCP-1 and glomerular filtration rate plus urinary sTNF-R1 and protein-creatinine ratio. Conclusion These results indicate that for active LN, sTNF-R1 could be a useful serum cytokine marker, with potential for VEGF in the urine. This study has confirmed the ability of the multiplex bead technique to detect cytokines in a good analytical range, including very low and high levels, in both serum and urine. Combining serum and urine markers provided additional sensitivity in distinguishing active from inactive LN.
Collapse
Affiliation(s)
- Z Adhya
- Immunology, King’s College Hospital, London, UK
- Immunology, Guy’s & St Thomas’ Hospitals, London, UK
| | - M El Anbari
- Research Branch, Sidra Medicine, Doha, Qatar
| | - S Anwar
- Section of Nephrology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - N Marr
- Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - M Y Karim
- Immunology, Guy’s & St Thomas’ Hospitals, London, UK
- Lupus Unit, Guy’s & St Thomas’ Hospitals, London, UK
- Pathology, Sidra Medicine, Doha, Qatar
| |
Collapse
|
41
|
Gasparin AA, Pamplona Bueno de Andrade N, Hax V, Tres GL, Veronese FV, Monticielo OA. Urinary biomarkers for lupus nephritis: the role of the vascular cell adhesion molecule-1. Lupus 2019; 28:265-272. [PMID: 30712490 DOI: 10.1177/0961203319826695] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Renal involvement is one of the main complications of systemic lupus erythematosus, causing a significant impact on patients' morbidity and mortality. Renal biopsy is still the gold standard of diagnosis, but it has many limitations. In this sense, several recent studies aim to identify biomarkers that not only predict disease activity and renal histology, but also lead to earlier treatment. In previous studies, the soluble vascular cell adhesion molecule-1 measured in urine showed a strong association with the presence of lupus nephritis, with clinical and histological activity indexes of the disease and with more severe renal lesions. This paper reviews the main urinary biomarkers of lupus nephritis that have been studied, with special emphasis on vascular cell adhesion molecule-1 results.
Collapse
Affiliation(s)
- A A Gasparin
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | - V Hax
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - G Leví Tres
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - F V Veronese
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - O A Monticielo
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
42
|
Liu S, Li N, Zhu Q, Zhu B, Wu T, Wang G, Liu S, Luo Q. Increased Serum MCP-1 Levels in Systemic Vasculitis Patients with Renal Involvement. J Interferon Cytokine Res 2018; 38:406-412. [PMID: 30230985 DOI: 10.1089/jir.2017.0140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Shasha Liu
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| | - Nanfang Li
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| | - Qing Zhu
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| | - Bin Zhu
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| | - Ting Wu
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| | - Guoliang Wang
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| | - Shanshan Liu
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| | - Qin Luo
- The Center for Hypertension of the People's Hospital of Xinjiang Uygur Autonomous Region, Hypertension Institute of Xinjiang China, Xinjiang, China
| |
Collapse
|
43
|
Ding Y, Nie LM, Pang Y, Wu WJ, Tan Y, Yu F, Zhao MH. Composite urinary biomarkers to predict pathological tubulointerstitial lesions in lupus nephritis. Lupus 2018; 27:1778-1789. [PMID: 30020021 DOI: 10.1177/0961203318788167] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective This study aimed to evaluate the clinical value of urinary biomarkers including kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and monocyte chemoattractant protein-1 (MCP-1) in lupus nephritis. Methods A total of 109 biopsy-proven lupus nephritis patients were included and 50 healthy individuals were used as normal controls. Urinary KIM-1, NGAL, and MCP-1 levels were measured by ELISA and their correlations with clinical and histological features were assessed. Receiver operating characteristic curves were performed and the Cox regression model was applied to identify prognostic factors associated with renal outcomes. Results Active lupus nephritis patients exhibited elevated urinary levels of KIM-1, NGAL, and MCP-1 compared with lupus nephritis patients in remission ( P < 0.001) and normal controls ( P < 0.001). The urinary KIM-1 level was correlated with pathological tubular atrophy ( r = 0.208, P < 0.05) and increased significantly in the presence of interstitial inflammatory lesions ( P = 0.031). Urinary KIM-1, NGAL, and MCP-1 levels were higher in patients with active tubulointerstitial lesions than in those with only chronic lesions ( P = 0.015, P = 0.230, and P = 0.086, respectively). A combination of KIM-1, NGAL, and MCP-1 was a good indicator for diagnosing active tubulointerstitial lesions (area under the curve: 0.796). The combination of KIM-1 and NGAL was identified as an independent risk factor for renal outcomes (hazard ratio = 7.491, P < 0.05). Conclusion Urinary KIM-1, NGAL, and MCP-1 levels were associated with kidney injury indices in lupus nephritis. The combination of the three biomarkers showed increased power in predicting tubulointerstitial lesions and renal outcomes.
Collapse
Affiliation(s)
- Y Ding
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China.,2 Department of Nephrology, Peking University International Hospital, Beijing, PR China
| | - L-M Nie
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China.,3 Renal Division, Department of Medicine, First Hospital of Shijiazhuang, Shijiazhuang, Hebei, PR, China
| | - Y Pang
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China.,4 Institute of Nephrology, Peking University, Beijing, PR China.,5 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China.,6 Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - W-J Wu
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China.,4 Institute of Nephrology, Peking University, Beijing, PR China.,5 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China.,6 Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China.,7 Peking-Tsinghua Center for Life Sciences; Beijing, PR China
| | - Y Tan
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China.,4 Institute of Nephrology, Peking University, Beijing, PR China.,5 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China.,6 Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - F Yu
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China.,2 Department of Nephrology, Peking University International Hospital, Beijing, PR China.,4 Institute of Nephrology, Peking University, Beijing, PR China.,5 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China.,6 Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China
| | - M-H Zhao
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, PR China.,4 Institute of Nephrology, Peking University, Beijing, PR China.,5 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, PR China.,6 Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, PR China.,7 Peking-Tsinghua Center for Life Sciences; Beijing, PR China
| |
Collapse
|
44
|
Wolf SJ, Theros J, Reed TJ, Liu J, Grigorova IL, Martínez-Colón G, Jacob CO, Hodgin JB, Kahlenberg JM. TLR7-Mediated Lupus Nephritis Is Independent of Type I IFN Signaling. THE JOURNAL OF IMMUNOLOGY 2018; 201:393-405. [PMID: 29884703 DOI: 10.4049/jimmunol.1701588] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/18/2018] [Indexed: 12/15/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease characterized by increased type I IFNs, autoantibodies, and inflammatory-mediated multiorgan damage. TLR7 activation is an important contributor to systemic lupus erythematosus pathogenesis, but the mechanisms by which type I IFNs participate in TLR7-driven pathologic conditions remain uncertain. In this study, we examined the requirement for type I IFNs in TLR7-stimulated lupus nephritis. Lupus-prone NZM2328, INZM (which lack a functional type I IFN receptor), and NZM2328 IL-1β-/- mice were treated at 10 wk of age on the right ear with R848 (TLR7 agonist) or control (DMSO). Autoantibody production and proteinuria were assessed throughout treatment. Multiorgan inflammation was assessed at the time of decline in health. Renal infiltrates and mRNA expression were also examined after 14 d of treatment. Both NZM2328 and INZM mice exhibited a decline in survival after 3-4 wk of R848 but not vehicle treatment. Development of splenomegaly and liver inflammation were dependent on type I IFN. Interestingly, autoantibody production, early renal infiltration of dendritic cells, upregulation of IL-1β, and lupus nephritis occurred independent of type I IFN signaling. Development of TLR7-driven lupus nephritis was not abolished by the deletion of IL-1β. Thus, although IFN-α is sufficient to induce nephritis acceleration, our data emphasize a critical role for IFN-independent signaling in TLR7-mediated lupus nephritis. Further, despite upregulation of IL-1β after TLR7 stimulation, deletion of IL-1β is not sufficient to reduce lupus nephritis development in this model.
Collapse
Affiliation(s)
- Sonya J Wolf
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.,Immunology Program, University of Michigan, Ann Arbor, MI 48109
| | - Jonathan Theros
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Tammi J Reed
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Jianhua Liu
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Irina L Grigorova
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| | | | - Chaim O Jacob
- University of Southern California Keck School of Medicine, Los Angeles, CA 90033; and
| | - Jeffrey B Hodgin
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
45
|
Iris M, Tsou PS, Sawalha AH. Caffeine inhibits STAT1 signaling and downregulates inflammatory pathways involved in autoimmunity. Clin Immunol 2018; 192:68-77. [PMID: 29678503 DOI: 10.1016/j.clim.2018.04.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/16/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023]
Abstract
Caffeine is a widely consumed pharmacologically active product. We focused on characterizing immunomodulatory effects of caffeine on peripheral blood mononuclear cells. Caffeine at high doses showed a robust downregulatory effect on cytokine activity and genes related to several autoimmune diseases including lupus and rheumatoid arthritis. Dose-dependent validation experiments showed downregulation at the mRNA levels of key inflammation-related genes including STAT1, TNF, IFNG, and PPARG. TNF and PPARG were suppressed even with the lowest caffeine dose tested, which corresponds to the serum concentration of caffeine after administration of one cup of coffee. Cytokine levels of IL-8, MIP-1β, IL-6, IFN-γ, GM-CSF, TNF, IL-2, IL-4, MCP-1, and IL-10 were decreased significantly with caffeine treatment. Upstream regulator analysis suggests that caffeine inhibits STAT1 signaling, which was confirmed by showing reduced phosphorylated STAT1 after caffeine treatment. Further studies exploring disease-modulating potential of caffeine in autoimmune diseases and further exploring the mechanisms involved are warranted.
Collapse
Affiliation(s)
- Merve Iris
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Marmara University, School of Medicine, Istanbul, Turkey
| | - Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
46
|
Gómez-Puerta JA, Ortiz-Reyes B, Urrego T, Vanegas-García AL, Muñoz CH, González LA, Cervera R, Vásquez G. Urinary neutrophil gelatinase-associated lipocalin and monocyte chemoattractant protein 1 as biomarkers for lupus nephritis in Colombian SLE patients. Lupus 2017; 27:637-646. [DOI: 10.1177/0961203317738226] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- J A Gómez-Puerta
- Grupo de Inmunología Celular e Inmunogenética, Universidad de Antioquia, Medellín, Colombia
- Grupo de Reumatología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - B Ortiz-Reyes
- Grupo de Inmunología Celular e Inmunogenética, Universidad de Antioquia, Medellín, Colombia
| | - T Urrego
- Grupo de Inmunología Celular e Inmunogenética, Universidad de Antioquia, Medellín, Colombia
| | - A L Vanegas-García
- Grupo de Reumatología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
- Hospital Universitario de San Vicente Fundación, Medellín, Colombia
| | - C H Muñoz
- Grupo de Reumatología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
- Hospital Universitario de San Vicente Fundación, Medellín, Colombia
| | - L A González
- Grupo de Reumatología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - R Cervera
- Department of Autoimmune Diseases, Hospital Clinic, Barcelona, Spain
| | - G Vásquez
- Grupo de Inmunología Celular e Inmunogenética, Universidad de Antioquia, Medellín, Colombia
- Grupo de Reumatología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|