1
|
Li X, Villanueva V, Jimenez V, Nguyen B, Chauhan NR, Khan SQ, Dorschner JM, Jensen MA, Alzahrani K, Wei H, Cimbaluk DJ, Wei DC, Jolly M, Lopez-Rodriguez D, Pineda SB, Barbosa A, Vazquez-Padron RI, Faridi HM, Reiser J, Niewold TB, Gupta V. CD11b suppresses TLR7-driven inflammatory signaling to protect against lupus nephritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605143. [PMID: 39211173 PMCID: PMC11361177 DOI: 10.1101/2024.07.26.605143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Lupus Nephritis (LN) is a severe complication of systemic lupus erythematosus (SLE) that affects kidney function. Here, we investigated the role of CD11b, a protein encoded by the ITGAM gene, in the development of LN and its functional activation as a therapeutic strategy. Genetic coding variants of ITGAM significantly increase the risk for SLE and LN by producing a less active CD11b and leading to elevated levels of type I interferon (IFN I). However, a molecular mechanism for how these variants increase LN risk has been unclear. Here, we determined that these variants also significantly associate with elevations in soluble urokinase plasminogen activator receptor (suPAR), a known biomarker linked to kidney disease, suggesting a novel molecular connection. Pharmacologic activation of CD11b with a novel, clinical-stage agonist ONT01 significantly suppressed suPAR production in myeloid cells and reduced systemic inflammation and kidney damage in multiple experimental models of LN. Importantly, delaying treatment with ONT01 until after disease onset also significantly reduced serum suPAR and inflammatory cytokines, and decreased immune complex deposition in the glomerulus, glomerulonephritis and albuminuria, suggesting that CD11b activation is therapeutic for LN. Genetic activation of CD11b via a gain-of-function CD11b mutation also showed complete protection from LN, whereas genetic deletion of CD11b worsened the disease in mice, providing further evidence of the role of CD11b activation in regulating LN. Finally, transfer of human LN PBMCs generated human LN like disease in mice that was significantly reduced by ONT01. Together, these data provide strong evidence that ONT01 mediated CD11b activation can therapeutically modulate TLR7-driven inflammation and protect against LN. These findings support clinical development of CD11b agonists as novel therapeutics for treating lupus nephritis in human patients.
Collapse
|
2
|
Deng AF, Wang FX, Wang SC, Zhang YZ, Bai L, Su JC. Bone-organ axes: bidirectional crosstalk. Mil Med Res 2024; 11:37. [PMID: 38867330 PMCID: PMC11167910 DOI: 10.1186/s40779-024-00540-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
In addition to its recognized role in providing structural support, bone plays a crucial role in maintaining the functionality and balance of various organs by secreting specific cytokines (also known as osteokines). This reciprocal influence extends to these organs modulating bone homeostasis and development, although this aspect has yet to be systematically reviewed. This review aims to elucidate this bidirectional crosstalk, with a particular focus on the role of osteokines. Additionally, it presents a unique compilation of evidence highlighting the critical function of extracellular vesicles (EVs) within bone-organ axes for the first time. Moreover, it explores the implications of this crosstalk for designing and implementing bone-on-chips and assembloids, underscoring the importance of comprehending these interactions for advancing physiologically relevant in vitro models. Consequently, this review establishes a robust theoretical foundation for preventing, diagnosing, and treating diseases related to the bone-organ axis from the perspective of cytokines, EVs, hormones, and metabolites.
Collapse
Affiliation(s)
- An-Fu Deng
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fu-Xiao Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Si-Cheng Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200444, China
| | - Ying-Ze Zhang
- Department of Orthopaedics, the Third Hospital of Hebei Medical University, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, 050051, China.
| | - Long Bai
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, Zhejiang, China.
| | - Jia-Can Su
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
3
|
Liu M, He P, Ye Z, Yang S, Zhang Y, Wu Q, Zhou C, Zhang Y, Hou FF, Qin X. Functional gastrointestinal disorders, mental health, genetic susceptibility, and incident chronic kidney disease. Chin Med J (Engl) 2024; 137:1088-1094. [PMID: 37668042 PMCID: PMC11062687 DOI: 10.1097/cm9.0000000000002805] [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: 02/02/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Whether functional gastrointestinal disorders (FGIDs) are associated with the long-term risk of chronic kidney disease (CKD) remains unclear. We aimed to investigate the prospective association of FGIDs with CKD and examine whether mental health mediated the association. METHODS About 416,258 participants without a prior CKD diagnosis enrolled in the UK Biobank between 2006 and 2010 were included. Participants with FGIDs (including irritable bowel syndrome [IBS], dyspepsia, and other functional intestinal disorders [FIDs; mainly composed of constipation]) were the exposure group, and non-FGID participants were the non-exposure group. The primary outcome was incident CKD, ascertained from hospital admission and death registry records. A Cox proportional hazard regression model was used to investigate the association between FGIDs and CKD, and the mediation analysis was performed to investigate the mediation proportions of mental health. RESULTS At baseline, 33,156 (8.0%) participants were diagnosed with FGIDs, including 21,060 (5.1%), 8262 (2.0%), and 6437 (1.6%) cases of IBS, dyspepsia, and other FIDs, respectively. During a mean follow-up period of 12.1 years, 11,001 (2.6%) participants developed CKD. FGIDs were significantly associated with a higher risk of incident CKD compared to the absence of FGIDs (hazard ratio [HR], 1.36; 95% confidence interval [CI], 1.28-1.44). Similar results were observed for IBS (HR, 1.27; 95% CI, 1.17-1.38), dyspepsia (HR, 1.30; 95% CI, 1.17-1.44), and other FIDs (HR, 1.60; 95% CI, 1.43-1.79). Mediation analyses suggested that the mental health score significantly mediated 9.05% of the association of FGIDs with incident CKD and 5.63-13.97% of the associations of FGID subtypes with CKD. Specifically, the positive associations of FGIDs and FGID subtypes with CKD were more pronounced in participants with a high genetic risk of CKD. CONCLUSION Participants with FGIDs had a higher risk of incident CKD, which was partly explained by mental health scores and was more pronounced in those with high genetic susceptibility to CKD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong 510515, China
| |
Collapse
|
4
|
Lee TH, Chen JJ, Wu CY, Lin TY, Hung SC, Yang HY. Immunosenescence, gut dysbiosis, and chronic kidney disease: Interplay and implications for clinical management. Biomed J 2024; 47:100638. [PMID: 37524304 PMCID: PMC10979181 DOI: 10.1016/j.bj.2023.100638] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023] Open
Abstract
Immunosenescence refers to the immune system changes observed in individuals over 50 years old, characterized by diminished immune response and chronic inflammation. Recent investigations have highlighted similar immune alterations in patients with reduced kidney function. The immune system and kidney function have been found to be closely interconnected. Studies have shown that as kidney function declines, both innate and adaptive immunity are affected. Chronic kidney disease (CKD) patients exhibit decreased levels of naive and regular T cells, as well as naive and memory B cells, while memory T cell counts increase. Furthermore, research suggests that CKD and end-stage kidney disease (ESKD) patients experience early thymic dysfunction and heightened homeostatic proliferation of naive T cells. In addition to reduced thymic T cell production, CKD patients display shorter telomeres in both CD4+ and CD8+ T cells. Declining kidney function induces uremic conditions, which alter the intestinal metabolic environment and promote pathogen overgrowth while reducing diversity. This dysbiosis-driven imbalance in the gut microbiota can result in elevated production of uremic toxins, which, in turn, enter the systemic circulation due to compromised gut barrier function under uremic conditions. The accumulation of gut-derived uremic toxins exacerbates local and systemic kidney inflammation. Immune-mediated kidney damage occurs due to the activation of immune cells in the intestine as a consequence of dysbiosis, leading to the production of cytokines and soluble urokinase-type plasminogen activator receptor (suPAR), thereby contributing to kidney inflammation. In this review, we delve into the fundamental mechanisms of immunosenescence in CKD, encompassing alterations in adaptive immunity, gut dysbiosis, and an overview of the clinical findings pertaining to immunosenescence.
Collapse
Affiliation(s)
- Tao Han Lee
- Nephrology Department, Chansn Hospital, Taoyuan, Taiwan
| | - Jia-Jin Chen
- Kidney Research Center, Nephrology Department, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chao-Yi Wu
- Division of Allergy, Asthma, And Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Yun Lin
- Division of Nephrology, Taipei Buddhist Tzu Chi General Hospital, Buddhist Tzu Chi University, Taipei, Taiwan
| | - Szu-Chun Hung
- Division of Nephrology, Taipei Buddhist Tzu Chi General Hospital, Buddhist Tzu Chi University, Taipei, Taiwan.
| | - Huang-Yu Yang
- Kidney Research Center, Nephrology Department, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
5
|
Kim EY, Dryer SE. Role of Formyl Peptide Receptors and β-Arrestin-1 in suPAR Signal Transduction in Mouse Podocytes: Interactions with αVβ3-Integrin. Cells 2024; 13:172. [PMID: 38247863 PMCID: PMC10814688 DOI: 10.3390/cells13020172] [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: 12/07/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
The soluble urokinase plasminogen activator receptor (suPAR) has been implicated in a wide range of pathological conditions including primary nephrotic syndromes and acute kidney injuries. suPAR can trigger transduction cascades in podocytes by outside-in activation of αVβ3-integrin, but there is evidence that the functional cell surface response element is actually a complex of different types of receptors, which may also include the receptor for advanced glycation end-products (RAGE) and formyl peptide receptors (FPRs). Here we observed that ROS accumulation and Src activation could be evoked by continuous 24 h exposure to either suPAR or the FPR agonist fMLF. Responses to suPAR and fMLF were completely blocked by either the FPR antagonist WRW4 or by the αV-integrin inhibitor cilengitide. Moreover, endogenous podocyte mouse Fpr1 co-immunoprecipitates with β3-integrin, suggesting that these receptors occur as a complex on the cell surface. suPAR- and fMLF-evoked activation of Src and ROS differed in time course. Thus, robust pertussis toxin (PTX)-sensitive responses were evoked by 60 min exposures to fMLF but not to suPAR. By contrast, responses to 24 h exposures to either suPAR or fMLF were PTX-resistant and were instead abolished by knockdown of β-arrestin-1 (BAR1). FPRs, integrins, and RAGE (along with various Toll-like receptors) can all function as pattern-recognition receptors that respond to "danger signals" associated with infections and tissue injury. The fact that podocytes express such a wide array of pattern-recognition receptors suggests that the glomerular filter is designed to change its function under certain conditions, possibly to facilitate clearance of toxic macromolecules.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA;
| | - Stuart E. Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA;
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
6
|
Ismail A, Hayek SS. Role of Soluble Urokinase-Type Plasminogen Activator Receptor in Cardiovascular Disease. Curr Cardiol Rep 2023; 25:1797-1810. [PMID: 37948017 DOI: 10.1007/s11886-023-01991-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
PURPOSE OF REVIEW Chronic inflammation is a major contributor to cardiovascular disease (CVD) risk. Soluble urokinase plasminogen activator receptor (suPAR) is an immune-derived glycoprotein that is strongly associated with atherosclerotic disease. This review summarizes evidence on suPAR's role in CVD pathogenesis and its potential as a prognostic indicator and therapeutic target. RECENT FINDINGS Clinical, genetic, and experimental evidence supports suPAR's role as a pathogenic factor in atherosclerosis. suPAR promotes atherosclerosis through modulation of monocyte activation and function. Clinically, elevated suPAR levels are linked to increased cardiovascular risk across diverse populations. Ongoing clinical trials are evaluating therapies targeting suPAR signaling. Current evidence positions suPAR as a regulator of myeloid cell function that contributes to vascular inflammation and subsequent cardiovascular events. Additional research is needed to determine whether suPAR measurement can improve CVD risk prediction and enable personalized management. Overall, suPAR is a promising immune-derived biomarker and target for reducing inflammation and cardiovascular risk.
Collapse
Affiliation(s)
- Anis Ismail
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, 48109, MI, USA
| | - Salim S Hayek
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, 48109, MI, USA.
| |
Collapse
|
7
|
Vallianou NG, Kounatidis D, Panagopoulos F, Evangelopoulos A, Stamatopoulos V, Papagiorgos A, Geladari E, Dalamaga M. Gut Microbiota and Its Role in the Brain-Gut-Kidney Axis in Hypertension. Curr Hypertens Rep 2023; 25:367-376. [PMID: 37632662 DOI: 10.1007/s11906-023-01263-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE OF REVIEW The role of the gut microbiota in modulating blood pressure is increasingly being recognized, currently. The purpose of this review is to summarize recent findings about the mechanisms involved in hypertension with regard to the phenomenon of "gut dysbiosis." RECENT FINDINGS Gut dysbiosis, i.e., the imbalance between the gut microbiota and the host, is characterized by a disruption of the tight junction proteins, such as occludins, claudins, and JAMs (junctional adhesion molecules), resulting in increased gut permeability or the so called "leaky gut." Due to the influence of genetic as well as environmental factors, various metabolites produced by the gut microbiota, such as indole and p-cresol, are increased. Thereby, uremic toxins, such as indoxyl sulfates and p-cresol sulfates, accumulate in the blood and the urine, causing damage in the podocytes and the tubular cells. In addition, immunological mechanisms are implicated as well. In particular, a switch from M2 macrophages to M1 macrophages, which produce pro-inflammatory cytokines, occurs. Moreover, a higher level of Th17 cells, releasing large amounts of interleukin-17 (IL-17), has been reported, when a diet rich in salt is consumed. Therefore, apart from the aggravation of uremic toxins, which may account for direct harmful effects on the kidney, there is inflammation not only in the gut, but in the kidneys as well. This crosstalk between the gut and the kidney is suggested to play a crucial role in hypertension. Notably, the brain is also implicated, with an increasing sympathetic output. The brain-gut-kidney axis seems to be deeply involved in the development of hypertension and chronic kidney disease (CKD). The notion that, by modulating the gut microbiota, we could regulate blood pressure is strongly supported by the current evidence. A healthy diet, low in animal protein and fat, and low in salt, together with the utilization of probiotics, prebiotics, synbiotics, or postbiotics, may contribute to our fight against hypertension.
Collapse
Affiliation(s)
| | | | - Fotis Panagopoulos
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | | | | | - Eleni Geladari
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece
| |
Collapse
|
8
|
Sommerer C, Müller-Krebs S, Nadal J, Schultheiss UT, Friedrich N, Nauck M, Schmid M, Nußhag C, Reiser J, Eckardt KU, Zeier M, Hayek SS. Prospective Cohort Study of Soluble Urokinase Plasminogen Activation Receptor and Cardiovascular Events in Patients With CKD. Kidney Int Rep 2023; 8:2265-2275. [PMID: 38025216 PMCID: PMC10658273 DOI: 10.1016/j.ekir.2023.08.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/28/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Soluble urokinase plasminogen activation receptor (suPAR) is an immune-derived pathogenic factor for kidney and atherosclerotic disease. Whether the association between suPAR and cardiovascular (CV) outcomes is dependent on the severity of underlying kidney disease is unclear. Methods We measured serum suPAR levels in 4994 participants (mean age 60 years; 60% men; 36% with diabetes mellitus; mean estimated glomerular filtration rate (eGFR) 49 ml/min per 1.73 m2, SD 18) of the German Chronic Kidney Disease (GCKD) cohort and examined its association with all-cause death, CV death, and major CV events (MACE) across the range of eGFR and urine albumin-to-creatinine ratio (UACR). Results The median suPAR level was 1771 pg/ml (interquartile range [IQR] 1447-2254 pg/ml). SuPAR levels were positively and independently correlated with age, eGFR, UACR, and parathyroid hormone levels. There were 573 deaths, including 190 CV deaths and 683 MACE events at a follow-up time of 6.5 years. In multivariable analyses, suPAR levels (log2) were associated with all-cause death (hazard ratio [HR] 1.36, 95% confidence interval [CI] 1.21-1.53), CV death (HR 1.27, 95% CI 1.03-1.57), and MACE (HR 1.13, 95% CI 1.00-1.28), and were not found to differ according to diabetes mellitus status, baseline eGFR, UACR, or parathyroid hormone levels. In mediation analysis, suPAR's direct effect on all-cause death, CV death, and MACE accounted for 77%, 67%, and 60% of the total effect, respectively; whereas the effect mediated through eGFR accounted for 23%, 34%, and 40%, respectively. Conclusion In a large cohort of individuals with chronic kidney disease (CKD), suPAR levels were associated with mortality and CV outcomes independently of indices of kidney function, consistent with its independent role in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Claudia Sommerer
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Sandra Müller-Krebs
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jennifer Nadal
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Ulla T. Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Medicine IV, Nephrology and Primary Care, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Christian Nußhag
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Zeier
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Salim S. Hayek
- Department of Medicine, Division of Cardiology, University of Michigan, Michigan, USA
| |
Collapse
|
9
|
Liu X, Wang X, Zhang P, Fang Y, Liu Y, Ding Y, Zhang W. Intestinal homeostasis in the gut-lung-kidney axis: a prospective therapeutic target in immune-related chronic kidney diseases. Front Immunol 2023; 14:1266792. [PMID: 38022571 PMCID: PMC10646503 DOI: 10.3389/fimmu.2023.1266792] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
In recent years, the role of intestinal homeostasis in health has received increasing interest, significantly improving our understanding of the complex pathophysiological interactions of the gut with other organs. Microbiota dysbiosis, impaired intestinal barrier, and aberrant intestinal immunity appear to contribute to the pathogenesis of immune-related chronic kidney diseases (CKD). Meanwhile, the relationship between the pathological changes in the respiratory tract (e.g., infection, fibrosis, granuloma) and immune-related CKD cannot be ignored. The present review aimed to elucidate the new underlying mechanism of immune-related CKD. The lungs may affect kidney function through intestinal mediation. Communication is believed to exist between the gut and lung microbiota across long physiological distances. Following the inhalation of various pathogenic factors (e.g., particulate matter 2.5 mum or less in diameter, pathogen) in the air through the mouth and nose, considering the anatomical connection between the nasopharynx and lungs, gut microbiome regulates oxidative stress and inflammatory states in the lungs and kidneys. Meanwhile, the intestine participates in the differentiation of T cells and promotes the migration of various immune cells to specific organs. This better explain the occurrence and progression of CKD caused by upper respiratory tract precursor infection and suggests the relationship between the lungs and kidney complications in some autoimmune diseases (e.g., anti-neutrophil cytoplasm antibodies -associated vasculitis, systemic lupus erythematosus). CKD can also affect the progression of lung diseases (e.g., acute respiratory distress syndrome and chronic obstructive pulmonary disease). We conclude that damage to the gut barrier appears to contribute to the development of immune-related CKD through gut-lung-kidney interplay, leading us to establish the gut-lung-kidney axis hypothesis. Further, we discuss possible therapeutic interventions and targets. For example, using prebiotics, probiotics, and laxatives (e.g., Rhubarb officinale) to regulate the gut ecology to alleviate oxidative stress, as well as improve the local immune system of the intestine and immune communication with the lungs and kidneys.
Collapse
Affiliation(s)
- Xinyin Liu
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- Department of Traditional Chinese Medicine, Jiande First People’s Hospital, Jiande, Hangzhou, China
| | - Xiaoran Wang
- Department of Nephrology, The First People’s Hospital of Hangzhou Lin’an District, Hangzhou, China
| | - Peipei Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yiwen Fang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanyan Liu
- Department of Geriatric, Zhejiang Aged Care Hospital, Hangzhou, China
| | - Yueyue Ding
- Department of Geriatric, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Wen Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
10
|
Alkhaleq HA, Karram T, Fokra A, Hamoud S, Kabala A, Abassi Z. The Protective Pathways Activated in Kidneys of αMUPA Transgenic Mice Following Ischemia\Reperfusion-Induced Acute Kidney Injury. Cells 2023; 12:2497. [PMID: 37887341 PMCID: PMC10605904 DOI: 10.3390/cells12202497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/12/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023] Open
Abstract
Despite the high prevalence of acute kidney injury (AKI), the therapeutic approaches for AKI are disappointing. This deficiency stems from the poor understanding of the pathogenesis of AKI. Recent studies demonstrate that αMUPA, alpha murine urokinase-type plasminogen activator (uPA) transgenic mice, display a cardioprotective pathway following myocardial ischemia. We hypothesize that these mice also possess protective renal pathways. Male and female αMUPA mice and their wild type were subjected to 30 min of bilateral ischemic AKI. Blood samples and kidneys were harvested 48 h following AKI for biomarkers of kidney function, renal injury, inflammatory response, and intracellular pathways sensing or responding to AKI. αMUPA mice, especially females, exhibited attenuated renal damage in response to AKI, as was evident from lower SCr and BUN, normal renal histology, and attenuated expression of NGAL and KIM-1. Notably, αMUPA females did not show a significant change in renal inflammatory and fibrotic markers following AKI as compared with wild-type (WT) mice and αMUPA males. Moreover, αMUPA female mice exhibited the lowest levels of renal apoptotic and autophagy markers during normal conditions and following AKI. αMUPA mice, especially the females, showed remarkable expression of PGC1α and eNOS following AKI. Furthermore, MUPA mice showed a significant elevation in renal leptin expression before and following AKI. Pretreatment of αMUPA with leptin-neutralizing antibodies prior to AKI abolished their resistance to AKI. Collectively, the kidneys of αMUPA mice, especially those of females, are less susceptible to ischemic I/R injury compared to WT mice, and this is due to nephroprotective actions mediated by the upregulation of leptin, eNOS, ACE2, and PGC1α along with impaired inflammatory, fibrotic, and autophagy processes.
Collapse
Affiliation(s)
- Heba Abd Alkhaleq
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
| | - Tony Karram
- Department of Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Ahmad Fokra
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
| | - Shadi Hamoud
- Internal Medicine, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Aviva Kabala
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
- Laboratory Medicine, Rambam Health Care Campus, Haifa 3109601, Israel
| |
Collapse
|
11
|
Zhu K, Mukherjee K, Wei C, Hayek SS, Collins A, Gu C, Corapi K, Altintas MM, Wang Y, Waikar SS, Bianco AC, Koch A, Tacke F, Reiser J, Sever S. The D2D3 form of uPAR acts as an immunotoxin and may cause diabetes and kidney disease. Sci Transl Med 2023; 15:eabq6492. [PMID: 37729431 DOI: 10.1126/scitranslmed.abq6492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is a risk factor for kidney diseases. In addition to suPAR, proteolysis of membrane-bound uPAR results in circulating D1 and D2D3 proteins. We showed that when exposed to a high-fat diet, transgenic mice expressing D2D3 protein developed progressive kidney disease marked by microalbuminuria, elevated serum creatinine, and glomerular hypertrophy. D2D3 transgenic mice also exhibited insulin-dependent diabetes mellitus evidenced by decreased levels of insulin and C-peptide, impaired glucose-stimulated insulin secretion, decreased pancreatic β cell mass, and high fasting blood glucose. Injection of anti-uPAR antibody restored β cell mass and function in D2D3 transgenic mice. At the cellular level, the D2D3 protein impaired β cell proliferation and inhibited the bioenergetics of β cells, leading to dysregulated cytoskeletal dynamics and subsequent impairment in the maturation and trafficking of insulin granules. D2D3 protein was predominantly detected in the sera of patients with nephropathy and insulin-dependent diabetes mellitus. These sera inhibited glucose-stimulated insulin release from human islets in a D2D3-dependent manner. Our study showed that D2D3 injures the kidney and pancreas and suggests that targeting this protein could provide a therapy for kidney diseases and insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- Ke Zhu
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kamalika Mukherjee
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Agnieszka Collins
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Changkyu Gu
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Kristin Corapi
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Yong Wang
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
| | - Sushrut S Waikar
- Section of Nephrology, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA 02129, USA
| | - Antonio C Bianco
- Division of Endocrinology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Alexander Koch
- Department of Gastroenterology, Metabolic Diseases and Internal Intensive Care Medicine, University Hospital Aachen, 52072 Aachen, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sanja Sever
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
12
|
Salfi G, Casiraghi F, Remuzzi G. Current understanding of the molecular mechanisms of circulating permeability factor in focal segmental glomerulosclerosis. Front Immunol 2023; 14:1247606. [PMID: 37795085 PMCID: PMC10546017 DOI: 10.3389/fimmu.2023.1247606] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
The pathogenetic mechanisms underlying the onset and the post-transplant recurrence of primary focal segmental glomerulosclerosis (FSGS) are complex and remain yet to be fully elucidated. However, a growing body of evidence emphasizes the pivotal role of the immune system in both initiating and perpetuating the disease. Extensive investigations, encompassing both experimental models and patient studies, have implicated T cells, B cells, and complement as crucial actors in the pathogenesis of primary FSGS, with various molecules being proposed as potential "circulating factors" contributing to the disease and its recurrence post kidney-transplantation. In this review, we critically assessed the existing literature to identify essential pathways for a comprehensive characterization of the pathogenesis of FSGS. Recent discoveries have shed further light on the intricate interplay between these mechanisms. We present an overview of the current understanding of the engagement of distinct molecules and immune cells in FSGS pathogenesis while highlighting critical knowledge gaps that require attention. A thorough characterization of these intricate immune mechanisms holds the potential to identify noninvasive biomarkers that can accurately identify patients at high risk of post-transplant recurrence. Such knowledge can pave the way for the development of targeted and personalized therapeutic approaches in the management of FSGS.
Collapse
Affiliation(s)
| | - Federica Casiraghi
- Istituto di Ricerche Farmacologiche Mario Negri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Bergamo, Italy
| | | |
Collapse
|
13
|
Xu X, Qu S, Zhang C, Zhang M, Qin W, Ren G, Bao H, Li L, Zen K, Liu Z. CD8 T Cell-Derived Exosomal miR-186-5p Elicits Renal Inflammation via Activating Tubular TLR7/8 Signal Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301492. [PMID: 37395441 PMCID: PMC10477851 DOI: 10.1002/advs.202301492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/27/2023] [Indexed: 07/04/2023]
Abstract
T cells play an important role in the development of focal segmental glomerulosclerosis (FSGS). The mechanism underlying such T cell-based kidney disease, however, remains elusive. Here the authors report that activated CD8 T cells elicit renal inflammation and tissue injury via releasing miR-186-5p-enriched exosomes. Continuing the cohort study identifying the correlation of plasma level of miR-186-5p with proteinuria in FSGS patients, it is demonstrated that circulating miR-186-5p is mainly derived from activated CD8 T cell exosomes. Renal miR-186-5p, which is markedly increased in FSGS patients and mice with adriamycin-induced renal injury, is mainly delivered by CD8 T cell exosomes. Depleting miR-186-5p strongly attenuates adriamycin-induced mouse renal injury. Supporting the function of exosomal miR-186-5p as a key circulating pathogenic factor, intravenous injection of miR-186-5p or miR-186-5p-containing T cell exosomes results in mouse renal inflammation and tissue injury. Tracing the injected T cell exosomes shows their preferential distribution in mouse renal tubules, not glomerulus. Mechanistically, miR-186-5p directly activates renal tubular TLR7/8 signal and initiates tubular cell apoptosis. Mutating the TLR7-binding sequence on miR-186-5p or deleting mouse TLR7 largely abolishes renal tubular injuries induced by miR-186-5p or adriamycin. These findings reveal a causative role of exosomal miR-186-5p in T cell-mediated renal dysfunction.
Collapse
Affiliation(s)
- Xiaodong Xu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Shuang Qu
- School of Life Science and TechnologyChina Pharmaceutical University639 Longmian AvenueNanjingJiangsu211198China
| | - Changming Zhang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Weisong Qin
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Guisheng Ren
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Hao Bao
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Limin Li
- School of Life Science and TechnologyChina Pharmaceutical University639 Longmian AvenueNanjingJiangsu211198China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing University School of Life SciencesNanjingJiangsu210093China
| | - Zhihong Liu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| |
Collapse
|
14
|
Wei C, Datta PK, Siegerist F, Li J, Yashwanth S, Koh KH, Kriho NW, Ismail A, Luo S, Fischer T, Amber KT, Cimbaluk D, Landay A, Endlich N, Rappaport J, Hayek SS, Reiser J. SuPAR mediates viral response proteinuria by rapidly changing podocyte function. Nat Commun 2023; 14:4414. [PMID: 37479685 PMCID: PMC10362037 DOI: 10.1038/s41467-023-40165-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023] Open
Abstract
Elevation in soluble urokinase receptor (suPAR) and proteinuria are common signs in patients with moderate to severe coronavirus disease 2019 (COVID-19). Here we characterize a new type of proteinuria originating as part of a viral response. Inoculation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes increased suPAR levels and glomerulopathy in African green monkeys. Using an engineered mouse model with high suPAR expression, inhaled variants of SARS-CoV-2 spike S1 protein elicite proteinuria that could be blocked by either suPAR antibody or SARS-CoV-2 vaccination. In a cohort of 1991 COVID-19 patients, suPAR levels exhibit a stepwise association with proteinuria in non-Omicron, but not in Omicron infections, supporting our findings of biophysical and functional differences between variants of SARS-CoV-2 spike S1 protein and their binding to podocyte integrins. These insights are not limited to SARS-CoV-2 and define viral response proteinuria (VRP) as an innate immune mechanism and co-activation of podocyte integrins.
Collapse
Affiliation(s)
- Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
| | - Prasun K Datta
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany
- NIPOKA GmbH, 17489, Greifswald, Germany
| | - Jing Li
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Sudhini Yashwanth
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kwi Hye Koh
- Morphic Therapeutic, Waltham, MA, 02451, USA
| | - Nicholas W Kriho
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shengyuan Luo
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Tracy Fischer
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Kyle T Amber
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Dermatology, Rush University Medical Center, Chicago, IL, USA
| | - David Cimbaluk
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Alan Landay
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany
- NIPOKA GmbH, 17489, Greifswald, Germany
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
15
|
Huang Y, Huang S, Zhuo X, Lin M. Predictive value of suPAR in AKI: a systematic review and meta-analysis. Clin Exp Nephrol 2023; 27:1-11. [PMID: 36469196 PMCID: PMC9734903 DOI: 10.1007/s10157-022-02300-2] [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: 07/01/2022] [Accepted: 11/18/2022] [Indexed: 12/07/2022]
Abstract
BACKGROUND Some clinical trials have shown that soluble urokinase-type plasminogen activator receptor (suPAR) has good predictive value for acute kidney injury (AKI), but there is still a lack of evidence-based proof. Therefore, we conducted this systematic review and meta-analysis to evaluate the predictive value of suPAR for AKI. METHODS Pubmed, EMBASE, Cochrane Library, and Web of Science databases were searched until December 2021 to obtain the literature on the prediction of suPAR for AKI. The quality of the included studies was assessed using the QUADAS-2 scoring system, and a bivariate random-effect model was used for the meta-analysis. The present study has been registered on PROSPERO (Registration No. CRD42022324978). RESULTS Seven articles were included, involving 2,319 patients, 635 of whom were AKI patients. The meta-analysis results showed that the combined sensitivity of suPAR in predicting AKI was 0.77 (95% CI 0.67-0.84); the specificity was 0.64 (95% CI 0.53-0.75); the odds ratio of diagnosis was 6 (95% CI 3-10); the pooled positive likelihood ratio was 2.2 (95% CI 1.6-2.9); the pooled negative likelihood ratio was 0.36 (95% CI 0.26-0.52); and the area under the summary receiver-operating characteristic (SROC) curve was 0.77 (95% CI 0.12~0.99). Deek's funnel plot suggested no potential publication bias among included studies. CONCLUSION suPAR is a valuable biomarker for the prediction of AKI with relatively high predictive accuracy, but its clinical application needs improvements. SuPAR should be considered as an indicator in the subsequent development of more effective predictive tools for AKI.
Collapse
Affiliation(s)
- Yan Huang
- Department of Clinical Laboratory, Foshan Hospital of Traditional Chinese Medicine, 6 Qinren Road, Chancheng District, Foshan City, 528000 Guangdong Province China
| | - Shengchun Huang
- Department of Clinical Laboratory, Foshan Hospital of Traditional Chinese Medicine, 6 Qinren Road, Chancheng District, Foshan City, 528000 Guangdong Province China
| | - Xueya Zhuo
- Department of Clinical Laboratory, Foshan Hospital of Traditional Chinese Medicine, 6 Qinren Road, Chancheng District, Foshan City, 528000 Guangdong Province China
| | - Mintao Lin
- Department of Clinical Laboratory, Foshan Hospital of Traditional Chinese Medicine, 6 Qinren Road, Chancheng District, Foshan City, 528000 Guangdong Province China
| |
Collapse
|
16
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
17
|
Hindy G, Tyrrell DJ, Vasbinder A, Wei C, Presswalla F, Wang H, Blakely P, Ozel AB, Graham S, Holton GH, Dowsett J, Fahed AC, Amadi KM, Erne GK, Tekmulla A, Ismail A, Launius C, Sotoodehnia N, Pankow JS, Thørner LW, Erikstrup C, Pedersen OB, Banasik K, Brunak S, Ullum H, Eugen-Olsen J, Ostrowski SR, Haas ME, Nielsen JB, Lotta LA, Engström G, Melander O, Orho-Melander M, Zhao L, Murthy VL, Pinsky DJ, Willer CJ, Heckbert SR, Reiser J, Goldstein DR, Desch KC, Hayek SS. Increased soluble urokinase plasminogen activator levels modulate monocyte function to promote atherosclerosis. J Clin Invest 2022; 132:e158788. [PMID: 36194491 PMCID: PMC9754000 DOI: 10.1172/jci158788] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/29/2022] [Indexed: 01/26/2023] Open
Abstract
People with kidney disease are disproportionately affected by atherosclerosis for unclear reasons. Soluble urokinase plasminogen activator receptor (suPAR) is an immune-derived mediator of kidney disease, levels of which are strongly associated with cardiovascular outcomes. We assessed suPAR's pathogenic involvement in atherosclerosis using epidemiologic, genetic, and experimental approaches. We found serum suPAR levels to be predictive of coronary artery calcification and cardiovascular events in 5,406 participants without known coronary disease. In a genome-wide association meta-analysis including over 25,000 individuals, we identified a missense variant in the plasminogen activator, urokinase receptor (PLAUR) gene (rs4760), confirmed experimentally to lead to higher suPAR levels. Mendelian randomization analysis in the UK Biobank using rs4760 indicated a causal association between genetically predicted suPAR levels and atherosclerotic phenotypes. In an experimental model of atherosclerosis, proprotein convertase subtilisin/kexin-9 (Pcsk9) transfection in mice overexpressing suPAR (suPARTg) led to substantially increased atherosclerotic plaques with necrotic cores and macrophage infiltration compared with those in WT mice, despite similar cholesterol levels. Prior to induction of atherosclerosis, aortas of suPARTg mice excreted higher levels of CCL2 and had higher monocyte counts compared with WT aortas. Aortic and circulating suPARTg monocytes exhibited a proinflammatory profile and enhanced chemotaxis. These findings characterize suPAR as a pathogenic factor for atherosclerosis acting at least partially through modulation of monocyte function.
Collapse
Affiliation(s)
- George Hindy
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Population Medicine, Qatar University College of Medicine, QU Health, Doha, Qatar
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Daniel J. Tyrrell
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Feriel Presswalla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hui Wang
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Graham
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace H. Holton
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph Dowsett
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Akl C. Fahed
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kingsley-Michael Amadi
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace K. Erne
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Annika Tekmulla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Launius
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Mary E. Haas
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Jonas B. Nielsen
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Luca A. Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | | | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Venkatesh L. Murthy
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David J. Pinsky
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cristen J. Willer
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Daniel R. Goldstein
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Karl C. Desch
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
18
|
Goodchild TT, Li Z, Lefer DJ. Soluble urokinase plasminogen activator receptor: from biomarker to active participant in atherosclerosis and cardiovascular disease. J Clin Invest 2022; 132:e165868. [PMID: 36519539 PMCID: PMC9754098 DOI: 10.1172/jci165868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis contributes to the majority of deaths related to cardiovascular disease (CVD). Recently, the nonspecific inflammatory biomarker soluble urokinase plasminogen activator receptor (suPAR) has shown prognostic value in patients with CVD; however, it remains unclear whether suPAR participates in the disease process. In this issue of the JCI, Hindy and colleagues report on their evaluation of a multi-ethnic cohort of over 5,000 participants without known CVD. High suPAR levels correlated with incident CVD and atherosclerosis. Genetic analysis revealed two variants associated with the suPAR-encoding gene (PLAUR) with higher plasma suPAR levels. Notably, a mouse model with high suPAR levels possessed aortic tissue with a proinflammatory phenotype, including monocytes with enhanced chemotaxis similar to that seen in atherogenesis. These findings suggest a causal relationship between suPAR and coronary artery calcification and have clinical implications that extend to inflammatory disorders beyond CVD.
Collapse
Affiliation(s)
- Traci T. Goodchild
- Cardiovascular Center of Excellence, LSU Health–New Orleans, New Orleans, Louisiana, USA
| | - Zhen Li
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David J. Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
19
|
Luo S, Vasbinder A, Du‐Fay‐de‐Lavallaz JM, Gomez JMD, Suboc T, Anderson E, Tekumulla A, Shadid H, Berlin H, Pan M, Azam TU, Khaleel I, Padalia K, Meloche C, O'Hayer P, Catalan T, Blakely P, Launius C, Amadi K, Pop‐Busui R, Loosen SH, Chalkias A, Tacke F, Giamarellos‐Bourboulis EJ, Altintas I, Eugen‐Olsen J, Williams KA, Volgman AS, Reiser J, Hayek SS. Soluble Urokinase Plasminogen Activator Receptor and Venous Thromboembolism in COVID-19. J Am Heart Assoc 2022; 11:e025198. [PMID: 35924778 PMCID: PMC9683642 DOI: 10.1161/jaha.122.025198] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Background Venous thromboembolism (VTE) contributes significantly to COVID-19 morbidity and mortality. The urokinase receptor system is involved in the regulation of coagulation. Levels of soluble urokinase plasminogen activator receptor (suPAR) reflect hyperinflammation and are strongly predictive of outcomes in COVID-19. Whether suPAR levels identify patients with COVID-19 at risk for VTE is unclear. Methods and Results We leveraged a multinational observational study of patients hospitalized for COVID-19 with suPAR and D-dimer levels measured on admission. In 1960 patients (mean age, 58 years; 57% men; 20% Black race), we assessed the association between suPAR and incident VTE (defined as pulmonary embolism or deep vein thrombosis) using logistic regression and Fine-Gray modeling, accounting for the competing risk of death. VTE occurred in 163 (8%) patients and was associated with higher suPAR and D-dimer levels. There was a positive association between suPAR and D-dimer (β=7.34; P=0.002). Adjusted for clinical covariables, including D-dimer, the odds of VTE were 168% higher comparing the third with first suPAR tertiles (adjusted odds ratio, 2.68 [95% CI, 1.51-4.75]; P<0.001). Findings were consistent when stratified by D-dimer levels and in survival analysis accounting for death as a competing risk. On the basis of predicted probabilities from random forest, a decision tree found the combined D-dimer <1 mg/L and suPAR <11 ng/mL cutoffs, identifying 41% of patients with only 3.6% VTE probability. Conclusions Higher suPAR was associated with incident VTE independently of D-dimer in patients hospitalized for COVID-19. Combining suPAR and D-dimer identified patients at low VTE risk. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT04818866.
Collapse
Affiliation(s)
- Shengyuan Luo
- Department of MedicineRush University Medical CenterChicagoIL
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | | | | | - Tisha Suboc
- Department of MedicineRush University Medical CenterChicagoIL
| | - Elizabeth Anderson
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Annika Tekumulla
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Husam Shadid
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Hanna Berlin
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Michael Pan
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Tariq U. Azam
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Ibrahim Khaleel
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Kishan Padalia
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Chelsea Meloche
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Patrick O'Hayer
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Tonimarie Catalan
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Christopher Launius
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Kingsley‐Michael Amadi
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Rodica Pop‐Busui
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Medical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Athanasios Chalkias
- Department of Anesthesiology, Faculty of MedicineUniversity of ThessalyLarisaGreece
- Outcomes Research ConsortiumClevelandOH
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Charité Mitte and Campus Virchow‐KlinikumCharité University Medicine BerlinBerlinGermany
| | | | - Izzet Altintas
- Department of Clinical ResearchCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Jesper Eugen‐Olsen
- Department of Clinical ResearchCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Kim A. Williams
- Department of Internal MedicineUniversity of Louisville School of MedicineLouisvilleKY
| | | | - Jochen Reiser
- Department of MedicineRush University Medical CenterChicagoIL
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| |
Collapse
|
20
|
Liao TH, Wu HC, Liao MT, Hu WC, Tsai KW, Lin CC, Lu KC. The Perspective of Vitamin D on suPAR-Related AKI in COVID-19. Int J Mol Sci 2022; 23:10725. [PMID: 36142634 PMCID: PMC9500944 DOI: 10.3390/ijms231810725] [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: 08/31/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed the lives of millions of people around the world. Severe vitamin D deficiency can increase the risk of death in people with COVID-19. There is growing evidence that acute kidney injury (AKI) is common in COVID-19 patients and is associated with poorer clinical outcomes. The kidney effects of SARS-CoV-2 are directly mediated by angiotensin 2-converting enzyme (ACE2) receptors. AKI is also caused by indirect causes such as the hypercoagulable state and microvascular thrombosis. The increased release of soluble urokinase-type plasminogen activator receptor (suPAR) from immature myeloid cells reduces plasminogen activation by the competitive inhibition of urokinase-type plasminogen activator, which results in low plasmin levels and a fibrinolytic state in COVID-19. Frequent hypercoagulability in critically ill patients with COVID-19 may exacerbate the severity of thrombosis. Versican expression in proximal tubular cells leads to the proliferation of interstitial fibroblasts through the C3a and suPAR pathways. Vitamin D attenuates the local expression of podocyte uPAR and decreases elevated circulating suPAR levels caused by systemic inflammation. This decrease preserves the function and structure of the glomerular barrier, thereby maintaining renal function. The attenuated hyperinflammatory state reduces complement activation, resulting in lower serum C3a levels. Vitamin D can also protect against COVID-19 by modulating innate and adaptive immunity, increasing ACE2 expression, and inhibiting the renin-angiotensin-aldosterone system. We hypothesized that by reducing suPAR levels, appropriate vitamin D supplementation could prevent the progression and reduce the severity of AKI in COVID-19 patients, although the data available require further elucidation.
Collapse
Affiliation(s)
- Tzu-Hsien Liao
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Hsien-Chang Wu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology and Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Ching-Chieh Lin
- Department of Chest Medicine, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
21
|
Bai Y, Li Y, Tang Z, Hu L, Jiang X, Chen J, Huang S, Wu K, Xu W, Chen C. Urinary proteome analysis of acute kidney injury in post-cardiac surgery patients using enrichment materials with high-resolution mass spectrometry. Front Bioeng Biotechnol 2022; 10:1002853. [PMID: 36177176 PMCID: PMC9513377 DOI: 10.3389/fbioe.2022.1002853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Cardiac surgery-associated acute kidney injury (CSA-AKI) may increase the mortality and incidence rates of chronic kidney disease in critically ill patients. This study aimed to investigate the underlying correlations between urinary proteomic changes and CSA-AKI. Methods: Nontargeted proteomics was performed using nano liquid chromatography coupled with Orbitrap Exploris mass spectrometry (MS) on urinary samples preoperatively and postoperatively collected from patients with CSA-AKI. Gemini C18 silica microspheres were used to separate and enrich trypsin-hydrolysed peptides under basic mobile phase conditions. Differential analysis was conducted to screen out urinary differential expressed proteins (DEPs) among patients with CSA-AKI for bioinformatics. Kyoto Encyclopedia of Genes and Genomes (KEGG) database analysis was adopted to identify the altered signal pathways associated with CSA-AKI. Results: Approximately 2000 urinary proteins were identified and quantified through data-independent acquisition MS, and 324 DEPs associated with AKI were screened by univariate statistics. According to KEGG enrichment analysis, the signal pathway of protein processing in the endoplasmic reticulum was enriched as the most up-regulated DEPs, and cell adhesion molecules were enriched as the most down-regulated DEPs. In protein–protein interaction analysis, the three hub targets in the up-regulated DEPs were α-1-antitrypsin, β-2-microglobulin and angiotensinogen, and the three key down-regulated DEPs were growth arrest-specific protein 6, matrix metalloproteinase-9 and urokinase-type plasminogen activator. Conclusion: Urinary protein disorder was observed in CSA-AKI due to ischaemia and reperfusion. The application of Gemini C18 silica microspheres can improve the protein identification rate to obtain highly valuable resources for the urinary DEPs of AKI. This work provides valuable knowledge about urinary proteome biomarkers and essential resources for further research on AKI.
Collapse
Affiliation(s)
- Yunpeng Bai
- Center of Scientific Research, Maoming People’s Hospital, Maoming, China
- Department of Critical Care Medicine, Maoming People’s Hospital, Maoming, China
| | - Ying Li
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhizhong Tang
- Department of Urology, Maoming People’s Hospital, Maoming, China
| | - Linhui Hu
- Department of Critical Care Medicine, Maoming People’s Hospital, Maoming, China
| | - Xinyi Jiang
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jingchun Chen
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Sumei Huang
- Center of Scientific Research, Maoming People’s Hospital, Maoming, China
- Department of Emergency, Maoming People’s Hospital, Maoming, China
- Biological Resource Center of Maoming People’s Hospital, Maoming, China
| | - Kunyong Wu
- Center of Scientific Research, Maoming People’s Hospital, Maoming, China
- Biological Resource Center of Maoming People’s Hospital, Maoming, China
| | - Wang Xu
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chunbo Chen
- Department of Emergency, Maoming People’s Hospital, Maoming, China
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Southern Medical University, Guangzhou, China
- *Correspondence: Chunbo Chen, , orcid.org/0000-0001-5662-497X
| |
Collapse
|
22
|
Abstract
Understanding the loss of kidney function resulting from kidney aging has become an emerging research focus that will facilitate the future development of antisenolytic treatments. In this issue of the JCI, Pippin et al. first identified PD-1 upregulation in the aged mouse podocyte via unbiased RNA-seq analysis. Overexpression of PD-1 in immortalized mouse podocytes induced cell death and a senescence-associated secretory phenotype, suggesting the pathological role of PD-1 upregulation in aged podocytes. Blocking PD-1 signaling via a neutralizing anti-PD-1 antibody reversed the aged phenotype in the aged mice and ameliorated proteinuria in an experimental focal segmental glomerulosclerosis (FSGS) mouse model. These findings highlight the role of PD-1 signaling in kidney aging and its therapeutic potential for human clinical trials.
Collapse
Affiliation(s)
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, and
- Department of Pharmacological Sciences, Mount Sinai School of Medicine, New York, New York, USA
- James J. Peters Veteran Administration Medical Center, New York, New York, USA
| |
Collapse
|
23
|
Skalec T, Adamik B, Kobylinska K, Gozdzik W. Soluble Urokinase-Type Plasminogen Activator Receptor Levels as a Predictor of Kidney Replacement Therapy in Septic Patients with Acute Kidney Injury: An Observational Study. J Clin Med 2022; 11:1717. [PMID: 35330042 PMCID: PMC8954771 DOI: 10.3390/jcm11061717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
The soluble urokinase-type plasminogen activator receptor (suPAR) is involved in the pathogenesis of acute kidney injury (AKI). Our goal was to establish the optimal suPAR cut-off point for predicting the need for kidney replacement therapy (KRT) use in sepsis patients and to analyze survival rates based on the suPAR level, AKI diagnosis, and the requirement for KRT. In total, 51 septic patients were included (82% septic shock; 96% mechanically ventilated, 35% KRT). Patients were stratified according to the AKI diagnosis and the need for KRT into three groups: AKI(+)/KRT(+), AKI(+)/KRT(−), and AKI(−)/KRT(−). A control group (N = 20) without sepsis and kidney failure was included. Sepsis patients had higher levels of the suPAR than control (13.01 vs. 4.05 ng/mL, p < 0.001). On ICU admission, the suPAR level was significantly higher in the AKI(+)/KRT(+) group than in the AKI(+)/KRT(−) and AKI(−)/KRT(−) groups (18.5 vs. 10.6 and 9.5 ng/mL, respectively; p = 0.001). The optimal suPAR cut-off point for predicting the need for KRT was established at 10.422 ng/mL (area under the curve 0.801, sensitivity 0.889, specificity 0.636). Moreover, patients AKI(+)/KRT(+) had the lowest probability of survival compared to patients AKI(+)/KRT(−) and AKI(−)/KRT(−) (p = 0.0003). The results indicate that the suPAR measurements may constitute an important element in the diagnosis of a patient with sepsis.
Collapse
Affiliation(s)
- Tomasz Skalec
- Clinical Department of Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Borowska St. 213, 50-556 Wroclaw, Poland; (T.S.); (W.G.)
| | - Barbara Adamik
- Clinical Department of Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Borowska St. 213, 50-556 Wroclaw, Poland; (T.S.); (W.G.)
| | - Katarzyna Kobylinska
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland;
| | - Waldemar Gozdzik
- Clinical Department of Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Borowska St. 213, 50-556 Wroclaw, Poland; (T.S.); (W.G.)
| |
Collapse
|
24
|
Vlasschaert C, Moran SM, Rauh MJ. The Myeloid-Kidney Interface in Health and Disease. Clin J Am Soc Nephrol 2022; 17:323-331. [PMID: 34507968 PMCID: PMC8823925 DOI: 10.2215/cjn.04120321] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Kidney homeostasis is highly dependent upon the correct functioning of myeloid cells. These cells form a distributed surveillance network throughout the kidney, where they play an integral role in the response to organ threat. Dysregulation of resident proinflammatory and profibrotic macrophages leads to kidney structural damage and scarring after kidney injury. Fibrosis throughout the kidney parenchyma contributes to the progressive functional decline observed in CKD, independent of the etiology. Circulating myeloid cells bearing intrinsic defects also affect the kidney substructures, such as neutrophils activated by autoantibodies that cause GN in ANCA-associated vasculitis. The kidney can also be affected by disorders of myelopoiesis, including myeloid leukemias (acute and chronic myeloid leukemias) and myelodysplastic syndromes. Clonal hematopoiesis of indeterminate potential is a common, newly recognized premalignant clinical entity characterized by clonal expansion of hyperinflammatory myeloid lineage cells that may have significant kidney sequelae. A number of existing therapies in CKD target myeloid cells and inflammation, including glucocorticoid receptor agonists and mineralocorticoid receptor antagonists. The therapeutic indications for these and other myeloid cell-targeted treatments is poised to expand as our understanding of the myeloid-kidney interface evolves.
Collapse
Affiliation(s)
| | - Sarah M. Moran
- Department of Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Michael J. Rauh
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada
| |
Collapse
|
25
|
Rasmussen LJH, Petersen JEV, Eugen-Olsen J. Soluble Urokinase Plasminogen Activator Receptor (suPAR) as a Biomarker of Systemic Chronic Inflammation. Front Immunol 2021; 12:780641. [PMID: 34925360 PMCID: PMC8674945 DOI: 10.3389/fimmu.2021.780641] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/09/2021] [Indexed: 01/08/2023] Open
Abstract
Systemic chronic inflammation (SCI) is persistent, health-damaging, low-grade inflammation that plays a major role in immunosenescence and in development and progression of many diseases. But currently, there are no recognized standard biomarkers to assess SCI levels alone, and SCI is typically measured by combining biomarkers of acute inflammation and infection, e.g., CRP, IL-6, and TNFα. In this review, we highlight 10 properties and characteristics that are shared by the blood protein soluble urokinase plasminogen activator receptor (suPAR) and SCI, supporting the argument that suPAR is a biomarker of SCI: (1) Expression and release of suPAR is upregulated by immune activation; (2) uPAR and suPAR exert pro-inflammatory functions; (3) suPAR is associated with the amount of circulating immune cells; (4) Blood suPAR levels correlate with the levels of established inflammatory biomarkers; (5) suPAR is minimally affected by acute changes and short-term influences, in contrast to many currently used markers of systemic inflammation; (6) Like SCI, suPAR is non-specifically associated with multiple diseases; (7) suPAR and SCI both predict morbidity and mortality; (8) suPAR and SCI share the same risk factors; (9) suPAR is associated with risk factors and outcomes of inflammation above and beyond other inflammatory biomarkers; (10) The suPAR level can be reduced by anti-inflammatory interventions and treatment of disease. Assessing SCI has the potential to inform risk for morbidity and mortality. Blood suPAR is a newer biomarker which may, in fact, be a biomarker of SCI since it is stably associated with inflammation and immune activation; shares the same risk factors as many age-related diseases; is both elevated by and predicts age-related diseases. There is strong evidence that suPAR is a prognostic marker of adverse events, morbidity, and mortality. It is associated with immune activity and prognosis across diverse conditions, including kidney disease, cardiovascular disease, cancer, diabetes, and inflammatory disorders. Thus, we think it likely represents a common underlying disease-process shared by many diseases; that is, SCI. We review the supporting literature and propose a research agenda that can help test the hypothesis that suPAR indexes SCI, with the potential of becoming the new gold standard for measuring SCI.
Collapse
Affiliation(s)
- Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| | - Jens Emil Vang Petersen
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| |
Collapse
|
26
|
Lupușoru G, Ailincăi I, Sorohan BM, Andronesi A, Achim C, Micu G, Caragheorgheopol A, Manda D, Lupușoru M, Ismail G. Serum soluble urokinase plasminogen activator receptor as a potential biomarker of renal impairment severity in diabetic nephropathy. Diabetes Res Clin Pract 2021; 182:109116. [PMID: 34728182 DOI: 10.1016/j.diabres.2021.109116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 12/28/2022]
Abstract
AIMS To investigate serum soluble form of urokinase-type plasminogen activator receptor (suPAR) in patients with diabetic kidney disease (DKD) and biopsy-proven diabetic nephropathy (DN), its correlation with histological parameters and its capacity as a biomarker for renal impairment severity. METHODS We conducted a cross-sectional study on 75 patients with diabetes mellitus (DM) and DKD, among whom 28 had biopsy-proven DN. RESULTS Among the 75 patients, 9 (12%) had type 1 and 66 (88%) type 2 DM. The median value of the serum suPAR level was 2857.2 pg/mL (1916.4-3700) in the entire cohort and 2472.1 pg/mL (1782.6-3745.8) in the biopsy-proven DN subgroup, respectively. suPAR was significantly correlated with diabetes duration, diabetic retinopathy, anti-proteinuric treatment, albuminuria, kidney function, DN class, interstitial fibrosis and tubular atrophy (IFTA) score and with interstitial inflammation score. suPAR had a good accuracy for the association with chronic kidney disease (CKD) stages G3b-5, macroalbuminuria, DN class IV, IFTA score 3 and interstitial inflammation score 2. CONCLUSIONS Serum suPAR was increased in DN patients and was associated with DM duration, diabetic retinopathy, renoprotective treatment, kidney function, proteinuria, DN class, IFTA and interstitial inflammation scores. Also, suPAR had a good capacity as a biomarker for advanced renal impairment and severe histological lesions of DN.
Collapse
Affiliation(s)
- Gabriela Lupușoru
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Ioana Ailincăi
- Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Bogdan Marian Sorohan
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania.
| | - Andreea Andronesi
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Camelia Achim
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Georgia Micu
- Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Andra Caragheorgheopol
- Research Department, "C.I.Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Dana Manda
- Research Department, "C.I.Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Mircea Lupușoru
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Gener Ismail
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
27
|
Lin JC, Chen XD, Xu ZR, Zheng LW, Chen ZH. Association of the Circulating Supar Levels with Inflammation, Fibrinolysis, and Outcome in Severe Burn Patients. Shock 2021; 56:948-955. [PMID: 34779798 PMCID: PMC8579993 DOI: 10.1097/shk.0000000000001806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hyperfibrinolysis and pro/anti-inflammatory imbalance usually occur in the early stage of severe burns. Soluble urokinase-type plasminogen activator receptor (suPAR) is involved in fibrinolysis and inflammation. To date, the levels of circulating suPAR in non-survivors with severe burns remain unknown. This study aimed to investigate the early association between circulating suPAR levels and biomarkers of fibrinolysis, pro/anti-inflammatory, and prognosis. METHODS Sixty-four consecutive Chinese patients with severe burns and 26 healthy volunteers were enrolled in a prospective observational cohort. Clinical characteristics and laboratory data were collected prospectively. Blood samples were collected at 48 h post-burn, and suPAR and biomarkers of pro/anti-inflammatory and fibrinolysis were detected by enzyme-linked immunosorbent assays. Important indicators between non-survivors and survivors were compared. Linear regression analysis was performed to screen variables associated with suPAR. Logistic regression analysis and receiver operating characteristic curve (ROC) analysis were performed to evaluate the prognostic value of suPAR. RESULT Compared with the control group, the circulating suPAR levels in the survivors (P < 0.001) and non-survivors (P = 0.017) were higher. Compared with survivors, non-survivors had lower circulating suPAR levels at 48 h post-burn, and they showed a higher degree of fibrinolysis (higher D-dimer) and a lower TNF-α/IL-10 ratio. According to linear regression analysis, the variables independently associated with a lower suPAR level were lower platelet factor 4 (PF-4), urokinase-type plasminogen activator (uPA), and TNF-α/IL-10 levels and a higher D-dimer level. Logistic regression and ROC analyses indicated that a suPAR level ≤ 4.70 μg/L was independently associated with 30-day mortality. CONCLUSION Low circulating suPAR levels at 48 h post-burn in severe burn patients may reflect decreased TNF-α/IL-10 ratio and increased hyperfibrinolysis. suPAR can predict 30-day mortality in patients with severe burn.
Collapse
Affiliation(s)
- Jian-Chang Lin
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Burn Institute, Fujian Burn Medical Center, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | | | | | | | | |
Collapse
|
28
|
Wang Y, Wu F, Chen C, Xu L, Lin W, Huang C, Yang Y, Wu S, Qi J, Cao H, Li G, Hong M, Zhu H. Soluble urokinase plasminogen activator receptor is associated with short-term mortality and enhanced reactive oxygen species production in acute-on-chronic liver failure. BMC Gastroenterol 2021; 21:429. [PMID: 34789156 PMCID: PMC8597314 DOI: 10.1186/s12876-021-02006-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 11/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute-on-chronic liver failure (ACLF) is a comprehensive syndrome characterized by an acute deterioration of liver function and high short-term mortality rates in patients with chronic liver disease. Whether plasma soluble urokinase plasminogen activator receptor (suPAR) is a suitable biomarker for the prognosis of patients with ACLF remains unknown. METHOD A prospective cohort of 282 patients with ACLF from three hospitals in China was included. 88.4% of the group was hepatitis B virus-related ACLF (HBV-related ACLF). Cox regression was used to assess the impact of plasma suPAR and other factors on 30- and 90-day mortality. Reactive oxygen species (ROS) production were detected to explore the role of suPAR in regulating neutrophil function in HBV-related ACLF. RESULT There was no difference in plasma suPAR levels between HBV-related and non-HBV-related ACLF. Patients with clinical complications had higher suPAR levels than those without these complications. A significant correlation was also found between suPAR and prognostic scores, infection indicators and inflammatory cytokines. Cox's regression multivariate analysis identified suPAR ≥ 14.7 ng/mL as a predictor for both day 30 and 90 mortality (Area under the ROC curve: 0.751 and 0.742 respectively), independent of the MELD and SOFA scores in patients with ACLF. Moreover, we firstly discovered suPAR enhanced neutrophil ROS production under E.coli stimulation in patients with HBV-related ACLF. CONCLUSIONS suPAR was a useful independent biomarker of short-term outcomes in patients with ACLF and might play a key role in the pathogenesis. Trial registration CNT, NCT02965560.
Collapse
Affiliation(s)
- Yunyun Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China.,Infectious Diseases Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fengtian Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Chao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Lichen Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Wei Lin
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunhong Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Ying Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Shanshan Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Jinjin Qi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Hanqin Cao
- Clinical Laboratory, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guojun Li
- Hepatology Department, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
| | - Meng Hong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China
| | - Haihong Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
29
|
Wei C, Spear R, Hahm E, Reiser J. suPAR, a Circulating Kidney Disease Factor. Front Med (Lausanne) 2021; 8:745838. [PMID: 34692736 PMCID: PMC8526732 DOI: 10.3389/fmed.2021.745838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023] Open
Abstract
Urokinase plasminogen activator receptor (uPAR) is a multifaceted, GPI-anchored three-domain protein. Release of the receptor results in variable levels of soluble uPAR (suPAR) in the blood circulation. suPAR levels have been linked to many disease states. In this mini-review, we discuss suPAR as a key circulating molecule mediating kidney disease with a particular focus on differently spliced isoforms.
Collapse
Affiliation(s)
- Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Ryan Spear
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Eunsil Hahm
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
30
|
Qin Y, Qiao Y, Wang D, Yan G, Tang C, Ma G. The Predictive Value of Soluble Urokinase-Type Plasminogen Activator Receptor in Contrast-Induced Acute Kidney Injury in Patients Undergoing Percutaneous Coronary Intervention. Int J Gen Med 2021; 14:6497-6504. [PMID: 34675617 PMCID: PMC8504866 DOI: 10.2147/ijgm.s339075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/01/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Soluble urokinase-type plasminogen activator receptor (SuPAR) is a circulating protein and a novel identified promising biomarker for various renal diseases and kidney injury. However, it remains unknown on the predictive value of suPAR in contrast induced acute kidney injury (CI-AKI) in patients undergoing percutaneous coronary intervention (PCI). Methods A total of 399 patients undergoing PCI were enrolled in the research from June 2020 to June 2021 in Zhongda Hospital. Patients were divided into CI-AKI and non-CI-AKI groups according to the preoperative and postoperative serum creatinine levels. Plasma suPAR level was detected through enzyme-linked immunosorbent assay on admission. Demographic data, hematological parameters, coronary angiography data and medications were recorded and compared between CI-AKI and non-CI-AKI groups. Logistic regression analysis and receiver operator characteristic (ROC) curve analysis were performed for identifying the independent risk factors of CI-AKI and assessment of the predictive value of suPAR for CI-AKI. Results CI-AKI occurred in 65 (16.3%) patients undergoing PCI. The level of suPAR in CI-AKI group was significantly higher than that in the non-CI-AKI group. Multivariate logistic regression indicated diabetes, lower LVEF, lower hydration rate, lower baseline eGFR, higher plasma suPAR (OR = 2.875, 95% CI = 2.038–3.672, P < 0.001) and volume of contrast media were all independent risk factors for CI-AKI after adjustment of the confounding factors. ROC analysis illustrated that the optimal area under the curve was 0.765, indicating plasma suPAR was a splendid predictor for CI-AKI. The corresponding cut-off value was 3.305 ng/mL, and the sensitivity and specificity were 63.1% and 82.3%, respectively. Conclusion Increased suPAR level is independently associated with elevated risk of suffering CI-AKI, and suPAR is a strong predictor for CI-AKI in patients undergoing PCI. SuPAR might act as a novel biomarker for CI-AKI in clinical practice.
Collapse
Affiliation(s)
- Yuhan Qin
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
31
|
Banu K, Lin Q, Basgen JM, Planoutene M, Wei C, Reghuvaran AC, Tian X, Shi H, Garzon F, Garzia A, Chun N, Cumpelik A, Santeusanio AD, Zhang W, Das B, Salem F, Li L, Ishibe S, Cantley LG, Kaufman L, Lemley KV, Ni Z, He JC, Murphy B, Menon MC. AMPK mediates regulation of glomerular volume and podocyte survival. JCI Insight 2021; 6:e150004. [PMID: 34473647 PMCID: PMC8525649 DOI: 10.1172/jci.insight.150004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 09/01/2021] [Indexed: 12/20/2022] Open
Abstract
Herein, we report that Shroom3 knockdown, via Fyn inhibition, induced albuminuria with foot process effacement (FPE) without focal segmental glomerulosclerosis (FSGS) or podocytopenia. Interestingly, knockdown mice had reduced podocyte volumes. Human minimal change disease (MCD), where podocyte Fyn inactivation was reported, also showed lower glomerular volumes than FSGS. We hypothesized that lower glomerular volume prevented the progression to podocytopenia. To test this hypothesis, we utilized unilateral and 5/6th nephrectomy models in Shroom3-KD mice. Knockdown mice exhibited less glomerular and podocyte hypertrophy after nephrectomy. FYN-knockdown podocytes had similar reductions in podocyte volume, implying that Fyn was downstream of Shroom3. Using SHROOM3 or FYN knockdown, we confirmed reduced podocyte protein content, along with significantly increased phosphorylated AMPK, a negative regulator of anabolism. AMPK activation resulted from increased cytoplasmic redistribution of LKB1 in podocytes. Inhibition of AMPK abolished the reduction in glomerular volume and induced podocytopenia in mice with FPE, suggesting a protective role for AMPK activation. In agreement with this, treatment of glomerular injury models with AMPK activators restricted glomerular volume, podocytopenia, and progression to FSGS. Glomerular transcriptomes from MCD biopsies also showed significant enrichment of Fyn inactivation and Ampk activation versus FSGS glomeruli. In summary, we demonstrated the important role of AMPK in glomerular volume regulation and podocyte survival. Our data suggest that AMPK activation adaptively regulates glomerular volume to prevent podocytopenia in the context of podocyte injury.
Collapse
Affiliation(s)
- Khadija Banu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Qisheng Lin
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - John M Basgen
- Morphometry and Stereology Laboratory, Charles R. Drew University of Medicine and Science, Los Angeles, California, USA
| | - Marina Planoutene
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anand C Reghuvaran
- Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xuefei Tian
- Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hongmei Shi
- Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Felipe Garzon
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aitor Garzia
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, New York, USA
| | - Nicholas Chun
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Arun Cumpelik
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew D Santeusanio
- Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bhaskar Das
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Li Li
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shuta Ishibe
- Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lloyd G Cantley
- Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lewis Kaufman
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kevin V Lemley
- Department of Pediatrics, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California, USA
| | - Zhaohui Ni
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Barbara Murphy
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Madhav C Menon
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
32
|
Agarwal S, Koh KH, Tardi NJ, Chen C, Dande RR, WerneckdeCastro JP, Sudhini YR, Luongo C, Salvatore D, Samelko B, Altintas MM, Mangos S, Bianco A, Reiser J. Deiodinase-3 is a thyrostat to regulate podocyte homeostasis. EBioMedicine 2021; 72:103617. [PMID: 34649077 PMCID: PMC8517284 DOI: 10.1016/j.ebiom.2021.103617] [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: 06/17/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Nephrotic syndrome (NS) is associated with kidney podocyte injury and may occur as part of thyroid autoimmunity such as Graves' disease. Therefore, the present study was designed to ascertain if and how podocytes respond to and regulate the input of biologically active thyroid hormone (TH), 3,5,3'-triiodothyronine (T3); and also to decipher the pathophysiological role of type 3 deiodinase (D3), a membrane-bound selenoenzyme that inactivates TH, in kidney disease. METHODS To study D3 function in healthy and injured (PAN, puromycin aminonucleoside and LPS, Lipopolysaccharide-mediated) podocytes, immunofluorescence, qPCR and podocyte-specific D3 knockout mouse were used. Surface plasmon resonance (SPR), co-immunoprecipitation and Proximity Ligation Assay (PLA) were used for the interaction studies. FINDINGS Healthy podocytes expressed D3 as the predominant deiodinase isoform. Upon podocyte injury, levels of Dio3 transcript and D3 protein were dramatically reduced both in vitro and in the LPS mouse model of podocyte damage. D3 was no longer directed to the cell membrane, it accumulated in the Golgi and nucleus instead. Further, depleting D3 from the mouse podocytes resulted in foot process effacement and proteinuria. Treatment of mouse podocytes with T3 phenocopied the absence of D3 and elicited activation of αvβ3 integrin signaling, which led to podocyte injury. We also confirmed presence of an active thyroid stimulating hormone receptor (TSH-R) on mouse podocytes, engagement and activation of which resulted in podocyte injury. INTERPRETATION The study provided a mechanistic insight into how D3-αvβ3 integrin interaction can minimize T3-dependent integrin activation, illustrating how D3 could act as a renoprotective thyrostat in podocytes. Further, injury caused by binding of TSH-R with TSH-R antibody, as found in patients with Graves' disease, explained a plausible link between thyroid disorder and NS. FUNDING This work was supported by American Thyroid Association (ATA-2018-050.R1).
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Kwi Hye Koh
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Nicholas J Tardi
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Chuang Chen
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | | | | | | | - Cristina Luongo
- Department of Public Health, University of Naples "Federico II," Naples, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples "Federico II," Naples, Italy
| | - Beata Samelko
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | | | - Steve Mangos
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Antonio Bianco
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, IL 60612.
| |
Collapse
|
33
|
Lanaret C, Anglicheau D, Audard V, Büchler M, Caillard S, Couzi L, Malvezzi P, Mesnard L, Bertrand D, Martinez F, Pernin V, Ducloux D, Poulain C, Thierry A, Del Bello A, Rerolle JP, Greze C, Uro-Coste C, Aniort J, Lambert C, Bouvier N, Schvartz B, Maillard N, Sayegh J, Oniszczuk J, Morin MP, Legendre C, Kamar N, Heng AE, Garrouste C. Rituximab for recurrence of primary focal segmental glomerulosclerosis after kidney transplantation: Results of a nationwide study. Am J Transplant 2021; 21:3021-3033. [PMID: 33512779 DOI: 10.1111/ajt.16504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 01/25/2023]
Abstract
Rituximab (RTX) therapy for primary focal segmental glomerulosclerosis recurrence after kidney transplantation (KT) has been extensively debated. We aimed to assess the benefit of adding RTX to plasmapheresis (PP), corticosteroids, and calcineurin inhibitors (standard of care, SOC). We identified 148 adult patients who received KT in 12/2004-12/2018 at 21 French centers: 109 received SOC (Group 1, G1), and 39 received immediate RTX along with SOC (Group 2, G2). In G1, RTX was introduced after 28 days of SOC in the event of failure (G1a, n = 19) or PP withdrawal (G1b, n = 12). Complete remission (CR) was achieved in 46.6% of patients, and partial remission (PR) was achieved in 33.1%. The 10-year graft survival rates were 64.7% and 17.9% in responders and nonresponders, respectively. Propensity score analysis showed no difference in CR+PR rates between G1 (82.6%) and G2 (71.8%) (p = .08). Following the addition of RTX (G1a), 26.3% of patients had CR, and 31.6% had PR. The incidence of severe infections was similar between patients treated with and without RTX. In multivariable analysis, infection episodes were associated with hypogammaglobulinemia <5 g/L. RTX could be used in cases of SOC failure or remission for early discontinuation of PP without increasing the risk of infection.
Collapse
Affiliation(s)
- Camille Lanaret
- Department of Nephrology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Dany Anglicheau
- Assistance Publique des Hôpitaux de Paris, Service de Néphrologie et Transplantation, Hôpital Universitaire Necker-Enfants Malades, Université de Paris, Paris, France
| | - Vincent Audard
- Assistance Publique des Hôpitaux de Paris (AP-HP, Service de Néphrologie et Transplantation Centre de Référence Maladie Rare «Syndrome Néphrotique Idiopathique», Hôpitaux Universitaires Henri-Mondor, Univ Paris Est Créteil, INSERM, IMRB, Créteil, France
| | - Mathias Büchler
- Service de Néphrologie et Immunologie Clinique, CHRU de Tours, Tours, France
| | - Sophie Caillard
- Service de Néphrologie, University Hospital, Strasbourg, France
| | - Lionel Couzi
- Service de Néphrologie, Transplantation, Dialyse et Aphérèses, CHU de Bordeaux, Bordeaux, France
| | - Paolo Malvezzi
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, CHU Grenoble-Alpes, Grenoble, France
| | - Laurent Mesnard
- Assistance Publique des Hôpitaux de Paris, Hôpital Universitaire Tenon, Urgences Néphrologiques et Transplantation Rénale, Université de Paris, Paris, France
| | | | - Franck Martinez
- Assistance Publique des Hôpitaux de Paris, Service de Néphrologie et Transplantation, Hôpital Universitaire Necker-Enfants Malades, Université de Paris, Paris, France
| | - Vincent Pernin
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Lapeyronie, CHU Montpellier, Montpellier, France
| | - Didier Ducloux
- Service de Néphrologie, Dialyse et Transplantation, CHU Besançon, Besançon, France
| | - Coralie Poulain
- Service de Néphrologie-Médecine Interne-Dialyse-Transplantation, CHU d'Amiens, Amiens, France
| | - Antoine Thierry
- Service de Néphrologie-Hémodialyse-Transplantation Rénale, CHU de Poitiers, Poitiers, France
| | - Arnaud Del Bello
- Département de Néphrologie et Transplantation d'Organes, CHU Toulouse, INSERM U1043, IFR-BMT, Université Paul Sabatier, Toulouse, France
| | - Jean P Rerolle
- Service de Néphrologie, Dialyse et Transplantation, CHU Limoges, Limoges, France
| | - Clarisse Greze
- Assistance Publique des Hôpitaux de Paris, Service de Néphrologie, Hôpital Universitaire Bichat-Claude-Bernard, Université de Paris, Paris, France
| | - Charlotte Uro-Coste
- Department of Nephrology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Julien Aniort
- Department of Nephrology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Céline Lambert
- Unité de Biostatistiques (DRCI, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Nicolas Bouvier
- Service de Néphrologie et Transplantation, CHU Caen, Caen, France
| | | | - Nicolas Maillard
- Service de Néphrologie et Transplantation, CHU Saint-Etienne, Saint-Etienne, France
| | - Johnny Sayegh
- Service de Néphrologie-Dialyse-Transplantation, CHU Angers, Angers, France
| | - Julie Oniszczuk
- Assistance Publique des Hôpitaux de Paris (AP-HP, Service de Néphrologie et Transplantation Centre de Référence Maladie Rare «Syndrome Néphrotique Idiopathique», Hôpitaux Universitaires Henri-Mondor, Univ Paris Est Créteil, INSERM, IMRB, Créteil, France
| | | | - Christophe Legendre
- Assistance Publique des Hôpitaux de Paris, Service de Néphrologie et Transplantation, Hôpital Universitaire Necker-Enfants Malades, Université de Paris, Paris, France
| | - Nassim Kamar
- Département de Néphrologie et Transplantation d'Organes, CHU Toulouse, INSERM U1043, IFR-BMT, Université Paul Sabatier, Toulouse, France
| | - Anne E Heng
- Department of Nephrology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Cyril Garrouste
- Department of Nephrology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| |
Collapse
|
34
|
Reisinger AC, Niedrist T, Posch F, Hatzl S, Hackl G, Prattes J, Schilcher G, Meißl AM, Raggam RB, Herrmann M, Eller P. Soluble urokinase plasminogen activator receptor (suPAR) predicts critical illness and kidney failure in patients admitted to the intensive care unit. Sci Rep 2021; 11:17476. [PMID: 34471146 PMCID: PMC8410930 DOI: 10.1038/s41598-021-96352-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is an inflammatory biomarker and risk factor for kidney diseases, with a potential prognostic value in critically ill patients. In this monocentric prospective study, we measured plasma suPAR levels immediately after ICU admission in unselected 237 consecutive patients using a turbidimetric assay. Primary objective was the prognostic value for ICU- and 28-day mortality. Secondary objectives were association with sequential organ failure assessment (SOFA) score, coagulation and inflammation markers, AKI-3 and differences in prespecified subgroups. Median suPAR levels were 8.0 ng/mL [25th-75th percentile 4.3-14.4], with lower levels in ICU survivors than non-survivors (6.7 vs. 11.6 ng/mL, p < 0.001). SuPAR levels were higher in COVID-19, kidney disease, moderate-to-severe liver disease, and sepsis. ICU mortality increased by an odds ratio (OR) of 4.7 in patients with the highest compared to lowest quartile suPAR. Kaplan-Meier overall survival estimates at 3 months were 63% and 49%, in patients with suPAR below/above 12 ng/mL (log-rank p = 0.027). Due to an observed interaction between SOFA score and suPAR, we performed a random forest method identifying cutoffs. ICU mortality was 53%, 17% and 2% in patients with a SOFA score > 7, SOFA ≤ 7 & suPAR > 8 ng/mL, and SOFA score ≤ 7 & suPAR ≤ 8 ng/mL, respectively. suPAR was a significant predictor for AKI-3 occurrence (OR per doubling 1.89, 95% CI: 1.20-2.98; p = 0.006). suPAR levels at ICU admission may offer additional value for risk stratification especially in ICU patients with moderate organ dysfunction as reflected by a SOFA score ≤ 7.
Collapse
Affiliation(s)
- Alexander C. Reisinger
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Tobias Niedrist
- grid.11598.340000 0000 8988 2476Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Florian Posch
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Stefan Hatzl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria ,grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Gerald Hackl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Juergen Prattes
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Section of Infectious Diseases and Tropical Medicine, Medical University of Graz, Graz, Austria
| | - Gernot Schilcher
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Anna-Maria Meißl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Reinhard B. Raggam
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Angiology, Medical University of Graz, Graz, Austria
| | - Markus Herrmann
- grid.11598.340000 0000 8988 2476Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| |
Collapse
|
35
|
Ouyang L, Su X, Li W, Tang L, Zhang M, Zhu Y, Xie C, Zhang P, Chen J, Huang H. ALKBH1-demethylated DNA N6-methyladenine modification triggers vascular calcification via osteogenic reprogramming in chronic kidney disease. J Clin Invest 2021; 131:146985. [PMID: 34003800 PMCID: PMC8279589 DOI: 10.1172/jci146985] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
Vascular calcification (VC) predicts cardiovascular morbidity and mortality in chronic kidney disease (CKD). To date, the underlying mechanisms remain unclear. We detected leukocyte DNA N6-methyladenine (6mA) levels in patients with CKD with or without aortic arch calcification. We used arteries from CKD mice infected with vascular smooth muscle cell-targeted (VSMC-targeted) adeno-associated virus encoding alkB homolog 1 (Alkbh1) gene or Alkbh1 shRNA to evaluate features of calcification. We identified that leukocyte 6mA levels were significantly reduced as the severity of VC increased in patients with CKD. Decreased 6mA demethylation resulted from the upregulation of ALKBH1. Here, ALKBH1 overexpression aggravated whereas its depletion blunted VC progression and osteogenic reprogramming in vivo and in vitro. Mechanistically, ALKBH1-demethylated DNA 6mA modification could facilitate the binding of octamer-binding transcription factor 4 (Oct4) to bone morphogenetic protein 2 (BMP2) promoter and activate BMP2 transcription. This resulted in osteogenic reprogramming of VSMCs and subsequent VC progression. Either BMP2 or Oct4 depletion alleviated the procalcifying effects of ALKBH1. This suggests that targeting ALKBH1 might be a therapeutic method to reduce the burden of VC in CKD.
Collapse
Affiliation(s)
- Liu Ouyang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaoyan Su
- Department of Nephropathy, Tungwah Hospital, Sun Yat-sen University, Dongguan, China
| | - Wenxin Li
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Liangqiu Tang
- Department of Cardiology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Mengbi Zhang
- Department of Nephropathy, Tungwah Hospital, Sun Yat-sen University, Dongguan, China
| | - Yongjun Zhu
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Changming Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Puhua Zhang
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Chen
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
36
|
Immune-mediated entities of (primary) focal segmental glomerulosclerosis. Cell Tissue Res 2021; 385:423-434. [PMID: 33907872 PMCID: PMC8523460 DOI: 10.1007/s00441-021-03454-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/19/2021] [Indexed: 12/21/2022]
Abstract
Focal segmental glomerulosclerosis (FSGS) represents a glomerular scar formation downstream of various different mechanisms leading to podocytopathy and podocyte loss. Recently, significant advances were made in understanding genetic factors, podocyte intrinsic mechanisms, and adaptive mechanisms causing FSGS. However, while most cases of nephrotic FSGS are being treated with immunosuppressants, the underlying immune dysregulation, involved immune cells, and soluble factors are only incompletely understood. Thus, we here summarize the current knowledge of proposed immune effector cells, secreted soluble factors, and podocyte response in immune-mediated (primary) FSGS.
Collapse
|
37
|
Loosen SH, Gorgulho J, Jördens MS, Schulze-Hagen M, Beier F, Vucur M, Schneider AT, Koppe C, Mertens A, Kather JN, Tacke F, Keitel V, Brümmendorf TH, Roderburg C, Luedde T. Serum Levels of Soluble Urokinase Plasminogen Activator Receptor Predict Tumor Response and Outcome to Immune Checkpoint Inhibitor Therapy. Front Oncol 2021; 11:646883. [PMID: 33869041 PMCID: PMC8047604 DOI: 10.3389/fonc.2021.646883] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/09/2021] [Indexed: 12/24/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have led to a paradigm shift in cancer therapy, improving outcomes in the treatment of various malignancies. However, not all patients benefit to the same extend from ICI. Reliable tools to predict treatment response and outcome are missing. Soluble urokinase plasminogen activator receptor (suPAR) is a marker of immune activation, whose levels are prognostic in various cancers. We evaluated circulating suPAR levels as a novel predictive and prognostic biomarker in patients receiving ICI therapy for solid tumors. Methods A total of n = 87 patients receiving ICI therapy for different solid malignancies as well as 32 healthy controls were included into this study. Serum levels of suPAR were measured by ELISA prior to and sequentially at two time points during ICI therapy. Results Baseline suPAR serum levels were significantly higher in solid tumor patients compared to healthy controls. Importantly, patients with low suPAR levels both before or during ICI treatment were more likely to have a favorable response to treatment at three and six months, respectively. This finding was confirmed by multivariate binary logistic regression analysis including several clinicopathological parameters. Moreover, circulating suPAR levels before and during therapy were an independent prognostic factor for overall survival (OS). As such, patients with initial suPAR levels above our ideal prognostic cut-off value (4.86 ng/ml) had a median OS of only 160 days compared to 705 days for patients with suPAR levels below this cut-off value. Finally, low baseline suPAR levels identified a subgroup of patients who experienced ICI-related side effects which in turn were associated with favorable treatment response and outcome. Conclusion Our data suggest that measurements of suPAR serum levels are a previously unknown, easily accessible tool to predict individual treatment response and outcome to ICI therapy. Circulating suPAR might therefore be implemented into stratification algorithms to identify the ideal candidates for ICI treatment.
Collapse
Affiliation(s)
- Sven H Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Joao Gorgulho
- Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany.,Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Markus S Jördens
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maximilian Schulze-Hagen
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Fabian Beier
- Department of Medicine IV, University Hospital RWTH Aachen, Aachen, Germany
| | - Mihael Vucur
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Anne T Schneider
- Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christiane Koppe
- Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Alexander Mertens
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jakob N Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tim H Brümmendorf
- Department of Medicine IV, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
38
|
Querfeld U. Cardiovascular disease in childhood and adolescence: Lessons from children with chronic kidney disease. Acta Paediatr 2021; 110:1125-1131. [PMID: 33080082 DOI: 10.1111/apa.15630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 11/26/2022]
Abstract
Children suffering from chronic kidney disease (CKD) have the apparent highest risk for the development of cardiovascular disease (CVD) at a young age. While symptoms of CVD are characteristically absent in childhood and adolescence, remodelling of the myocardium, medium and large-sized arteries and of the microcirculation is clinically significant and can be assessed with non-invasive technology. Kidney disease and its progression are the driver of CVD, mediated by an unparalleled accumulation of risk factors converging on several comorbid conditions including hypertension, anaemia, dyslipidaemia, disturbed mineral metabolism and chronic persistent inflammation. Large prospective paediatric cohorts studies have provided valuable insights into the pathogenesis and the progression of CKD-induced cardiovascular comorbidity and have characterised the cardiovascular phenotype in young patients. They have also provided the rationale for close monitoring of risk factors and have defined therapeutic targets. Recently discovered new biomarkers could help identify the individual risk for CVD. Prevention of CVD by aggressive therapy of modifiable risk factors is essential to enable long-term survival of young patients with CKD.
Collapse
Affiliation(s)
- Uwe Querfeld
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| |
Collapse
|
39
|
Jehn U, Schütte-Nütgen K, Henke U, Pavenstädt H, Suwelack B, Reuter S. Soluble urokinase-type plasminogen activator receptor (suPAR) is a risk indicator for eGFR loss in kidney transplant recipients. Sci Rep 2021; 11:3713. [PMID: 33580120 PMCID: PMC7880993 DOI: 10.1038/s41598-021-83333-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/02/2021] [Indexed: 12/03/2022] Open
Abstract
The prognostic significance of suPAR in various kidney diseases has recently been demonstrated. Its role in transplantation-specific outcomes is still largely unknown. Therefore, we prospectively investigated the prognostic relevance of suPAR in patients before and one year after kidney transplantation (KTx). We included 100 patients who had received a kidney transplantation between 2013 and 2015. The plasma concentration of suPAR was measured by ELISA assay. In recipients of living donations (LD), pre-transplant suPAR levels were significantly lower than those of recipients of deceased donations (DD). After KTx, suPAR levels significantly declined in LD and DD recipients, without a detectable difference between both groups any more. Higher suPAR levels in recipients one year after KTx were associated with a more severe eGFR loss in the following three years in multivariable cox-regression (n = 82, p = 0.021). suPAR-levels above 6212 pg/ml one year after KTx are associated with eGFR loss > 30%, which occurred almost twice as fast as in patients with suPAR ≤ 6212 pg/ml (p < 0.001). Hence, suPAR level at one year mark might be a risk indicator of increased eGFR loss.
Collapse
Affiliation(s)
- Ulrich Jehn
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, 48149, Muenster, Germany.
| | - Katharina Schütte-Nütgen
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, 48149, Muenster, Germany
| | - Ute Henke
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, 48149, Muenster, Germany
| | - Hermann Pavenstädt
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, 48149, Muenster, Germany
| | - Barbara Suwelack
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, 48149, Muenster, Germany
| | - Stefan Reuter
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, 48149, Muenster, Germany
| |
Collapse
|
40
|
Höbaus C, Ursli M, Yussef SM, Wrba T, Koppensteiner R, Schernthaner GH. Soluble urokinase-type plasminogen activator receptor predicts peripheral artery disease severity and outcomes. Vasc Med 2021; 26:11-17. [PMID: 33448911 PMCID: PMC7879231 DOI: 10.1177/1358863x20982077] [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] [Indexed: 11/25/2022]
Abstract
Soluble urokinase-type plasminogen activator receptor (suPAR) is associated with chronic kidney disease (CKD) severity and peripheral artery disease (PAD). We hypothesize an association of PAD severity and suPAR in patients without advanced CKD and further risk stratification according to the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines. For study purposes, suPAR was measured in 334 PAD patients (34% women, age 69 (62-78) years, eGFR 68 ± 20 mL/min/1.72 m2) by commercial ELISA. Patients were followed for 10 years to assess long-term all-cause survival by Cox regression. Higher suPAR levels were associated with lower ankle-brachial index (R = -0.215, p = 0.001) in patients with PAD without media-sclerosis (n = 236). suPAR levels inversely correlated with decreased glomerular filtration rate (R = -0.476, p < 0.001) and directly correlated with urinary albumin-to-creatinine ratio (R = 0.207, p < 0.001). Furthermore, higher suPAR levels associated with a higher KDIGO risk score (p < 0.001). Baseline suPAR was significantly associated with all-cause mortality (HR 1.40 (95% CI 1.16-1.68), p < 0.001) over 10 years. suPAR remained associated with mortality (HR 1.29 (1.03-1.61), p = 0.026) after multivariable adjustment for age, sex, cardiovascular risk factors, and eGFR. Future research may define a standard role for suPAR assessment in PAD's work-up and treatment, especially in patients with CKD.
Collapse
Affiliation(s)
- Clemens Höbaus
- Division of Angiology, Medicine II, Medical University Vienna, Vienna, Austria
| | - Martin Ursli
- Department of Internal Medicine I, University Hospital of St Poelten, Karl Landsteiner University of Health Sciences, Karl Landsteiner Institute for Nephrology and Hematooncology, St Poelten, Austria
| | | | - Thomas Wrba
- IT4Science, IT-Systems & Communications, Medical University Vienna, Vienna, Austria
| | | | | |
Collapse
|
41
|
Napolitano F, Montuori N. The Role of the Plasminogen Activation System in Angioedema: Novel Insights on the Pathogenesis. J Clin Med 2021; 10:518. [PMID: 33535668 PMCID: PMC7867209 DOI: 10.3390/jcm10030518] [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: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
The main physiological functions of plasmin, the active form of its proenzyme plasminogen, are blood clot fibrinolysis and restoration of normal blood flow. The plasminogen activation (PA) system includes urokinase-type plasminogen activator (uPA), tissue-type PA (tPA), and two types of plasminogen activator inhibitors (PAI-1 and PAI-2). In addition to the regulation of fibrinolysis, the PA system plays an important role in other biological processes, which include degradation of extracellular matrix such as embryogenesis, cell migration, tissue remodeling, wound healing, angiogenesis, inflammation, and immune response. Recently, the link between PA system and angioedema has been a subject of scientific debate. Angioedema is defined as localized and self-limiting edema of subcutaneous and submucosal tissues, mediated by bradykinin and mast cell mediators. Different forms of angioedema are linked to uncontrolled activation of coagulation and fibrinolysis systems. Moreover, plasmin itself can induce a potentiation of bradykinin production with consequent swelling episodes. The number of studies investigating the PA system involvement in angioedema has grown in recent years, highlighting its relevance in etiopathogenesis. In this review, we present the components and diverse functions of the PA system in physiology and its importance in angioedema pathogenesis.
Collapse
Affiliation(s)
| | - Nunzia Montuori
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80135 Naples, Italy;
| |
Collapse
|
42
|
Yuan C, Guo Z, Yu S, Jiang L, Huang M. Development of inhibitors for uPAR: blocking the interaction of uPAR with its partners. Drug Discov Today 2021; 26:1076-1085. [PMID: 33486111 DOI: 10.1016/j.drudis.2021.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/11/2021] [Indexed: 12/25/2022]
Abstract
Urokinase-type plasminogen activator receptor (uPAR) mediates a multitude of biological activities, has key roles in several clinical indications, including malignancies and inflammation, and, thus, has attracted intensive research over the past few decades. The pleiotropic functions of uPAR can be attributed to its interaction with an array of partners. Many inhibitors have been developed to intervene with the interaction of uPAR with these partners. Here, we review the development of these classes of uPAR inhibitor and their inhibitory mechanisms to promote the translation of these inhibitors to clinical applications.
Collapse
Affiliation(s)
- Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Zhanzhi Guo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fujian, 350116, China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fujian, 350116, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fujian, 350116, China.
| |
Collapse
|
43
|
Belliere J, Colombat M, Kounde C, Recher C, Ribes D, Huart A, Chauveau D, Demas V, Luquet I, Beyne-Rauzy O, Tavitian S, Faguer S. Kidney Involvement in Patients With Chronic Myelomonocytic Leukemia or BCR-ABL-Negative Myeloproliferative Neoplasms. Kidney Int Rep 2020; 6:737-745. [PMID: 33732988 PMCID: PMC7938079 DOI: 10.1016/j.ekir.2020.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 01/11/2023] Open
Abstract
Introduction The identification of specific molecular signatures and the development of new targeted drugs have changed the paradigm of onco-nephrology, now allowing a multiscale approach of kidney involvement related to hematologic malignancies relying on combined hematologic and molecular assessments. In this study, we aimed to refine the spectrum of kidney disorders associated with chronic myelomonocytic leukemia (CMML) or BCR-ABL–negative myeloproliferative neoplasms (MPNs), 2 very rare conditions scarcely described. Methods Case series. Patients with myeloid neoplasms who were referred to Toulouse University Hospital Nephrology Unit and were diagnosed with acute kidney injury (AKI), chronic kidney disease (CKD), or urine abnormalities were retrospectively included. Results Eighteen patients (males n=13, CMML n=8, essential thrombocytosis [ET] n=7, polycythemia vera [PV] n=1, and myelofibrosis n=2) developed kidney disease 7.7±2 years after the diagnosis of the malignancy. Twelve patients had AKI at presentation. Eight patients had glomerular presentation (high-range proteinuria 33%, microscopic hematuria 56%). Kidney biopsy (n=14) showed various patterns, including pauci-immune glomerulosclerosis (n=5), extramedullary hematopoiesis (n=6), or tubular atrophy and interstitial fibrosis with polymorphic inflammation (n=8). Immunostaining of CD61 confirmed the infiltration of megakaryocytes within glomeruli or interstitium in 5 of 8 patients. Other pictures of glomerulopathy were identified in 3 patients (IgA nephropathy n=2, AA amyloidosis n=1). Massive kidney infiltration by CMML was identified in 1 patient. After a mean follow-up of 24±6 months, malignancy was considered as stable in 11 patients (61%), but 22% of patients had progressed to end-stage renal failure. The remaining had persistently reduced kidney function. No correlation between the malignancy and the renal presentation and outcomes could be identified. Conclusions Kidney complications of CMML/MPN are heterogenous, and kidney biopsy may help to identify new molecular targets to prevent the development of kidney fibrosis.
Collapse
Affiliation(s)
- Julie Belliere
- Centre Hospitalier Universitaire de Toulouse, Département de Néphrologie et Transplantation d'Organes, Centre de reference des Maladies rénales rares, Toulouse, France.,INSERM U1048, Institut des maladies métaboliques et cardio-vasculaires, Toulouse, France.,Université Paul Sabatier, Toulouse, France
| | - Magali Colombat
- Université Paul Sabatier, Toulouse, France.,Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Département d'Anatomopathologie, Toulouse, France
| | - Clément Kounde
- Centre Hospitalier Universitaire de Toulouse, Département de Néphrologie et Transplantation d'Organes, Centre de reference des Maladies rénales rares, Toulouse, France
| | - Christian Recher
- Université Paul Sabatier, Toulouse, France.,Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Service d'Hématologie, Toulouse, France
| | - David Ribes
- Centre Hospitalier Universitaire de Toulouse, Département de Néphrologie et Transplantation d'Organes, Centre de reference des Maladies rénales rares, Toulouse, France
| | - Antoine Huart
- Centre Hospitalier Universitaire de Toulouse, Département de Néphrologie et Transplantation d'Organes, Centre de reference des Maladies rénales rares, Toulouse, France
| | - Dominique Chauveau
- Centre Hospitalier Universitaire de Toulouse, Département de Néphrologie et Transplantation d'Organes, Centre de reference des Maladies rénales rares, Toulouse, France.,INSERM U1048, Institut des maladies métaboliques et cardio-vasculaires, Toulouse, France.,Université Paul Sabatier, Toulouse, France
| | - Véronique Demas
- Université Paul Sabatier, Toulouse, France.,Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratoire d'Hématologie, Toulouse, France
| | - Isabelle Luquet
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratoire d'Hématologie, Toulouse, France
| | - Odile Beyne-Rauzy
- Université Paul Sabatier, Toulouse, France.,Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Service de Médecine interne, Toulouse, France
| | - Suzanne Tavitian
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Service d'Hématologie, Toulouse, France
| | - Stanislas Faguer
- Centre Hospitalier Universitaire de Toulouse, Département de Néphrologie et Transplantation d'Organes, Centre de reference des Maladies rénales rares, Toulouse, France.,INSERM U1048, Institut des maladies métaboliques et cardio-vasculaires, Toulouse, France.,Université Paul Sabatier, Toulouse, France
| |
Collapse
|
44
|
Raptis V, Loutradis C, Boutou AK, Faitatzidou D, Sioulis A, Ferro CJ, Papagianni A, Sarafidis PA. Serum Copeptin, NLPR3, and suPAR Levels among Patients with Autosomal-Dominant Polycystic Kidney Disease with and without Impaired Renal Function. Cardiorenal Med 2020; 10:440-451. [PMID: 33202410 DOI: 10.1159/000510834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/07/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The pathophysiology of renal disease progression in autosomal-dominant polycystic kidney disease (ADPKD) involves not only cystogenesis but also endothelial dysfunction, leading to the activation of inflammatory and fibrotic pathways. This study evaluated the levels of biomarkers related to osmoregulation, immune system activation, and tubular injury in ADPKD patients with impaired or preserved renal function. METHODS This study included 26 ADPKD patients with modestly impaired renal function (estimated glomerular filtration rate [eGFR] 45-70 mL/min/1.73 m2; Group A), 26 age- and sex-matched ADPKD patients with relatively preserved renal function (eGFR >70 mL/min/1.73 m2; Group B), and 26 age- and sex-matched controls (Group C). Serum levels of copeptin, the inflammasome nucleotide-binding and oligomerization domain-like receptors pyrin domain-containing protein 3 (NLRP3), and soluble urokinase-type plasminogen activator receptor (suPAR) were measured with ELISA techniques. RESULTS Patients in Group A had higher levels of copeptin (median [interquartile range]: 50.44 [334.85] pg/mL), NLRP3 (5.86 [3.89] ng/mL), and suPAR (390.05 [476.53] pg/mL) compared to patients in Group B (32.38 [58.33], p = 0.042; 2.42 [1.96], p < 0.001; and 313.78 [178.85], p = 0.035, respectively) and Group C (6.75 [6.43]; 1.09 [0.56]; and 198.30 [28.53], respectively; p < 0.001 for all comparisons). Levels of all studied markers were also significantly higher in Group B patients compared to controls (p < 0.001), despite having similar eGFR. In patients with ADPKD, all studied biomarkers were correlated positively with asymmetric-dimethylarginine (ADMA) and endocan levels, and negatively with eGFR. ADMA and endocan levels were the only parameters independently associated with increased copeptin levels. CONCLUSIONS This study showed that ADPKD patients with impaired and preserved renal function had higher copeptin, NLRP3, and suPAR levels than controls. Such findings support that cystogenesis and inflammation are associated with endothelial dysfunction, even in the early stages of ADKPD.
Collapse
Affiliation(s)
- Vasileios Raptis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charalampos Loutradis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Afroditi K Boutou
- Department of Respiratory Medicine, Papanikolaou General Hospital, Thessaloniki, Greece
| | - Danai Faitatzidou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Sioulis
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Charles J Ferro
- Department of Renal Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Aikaterini Papagianni
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pantelis A Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece,
| |
Collapse
|
45
|
Azam TU, Shadid HR, Blakely P, O'Hayer P, Berlin H, Pan M, Zhao P, Zhao L, Pennathur S, Pop-Busui R, Altintas I, Tingleff J, Stauning MA, Andersen O, Adami ME, Solomonidi N, Tsilika M, Tober-Lau P, Arnaoutoglou E, Keitel V, Tacke F, Chalkias A, Loosen SH, Giamarellos-Bourboulis EJ, Eugen-Olsen J, Reiser J, Hayek SS. Soluble Urokinase Receptor (SuPAR) in COVID-19-Related AKI. J Am Soc Nephrol 2020; 31:2725-2735. [PMID: 32963090 PMCID: PMC7608953 DOI: 10.1681/asn.2020060829] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AKI commonly occurs in patients with coronavirus disease 2019 (COVID-19). Its pathogenesis is poorly understood. The urokinase receptor system is a key regulator of the intersection between inflammation, immunity, and coagulation, and soluble urokinase plasminogen activator receptor (suPAR) has been identified as an immunologic risk factor for AKI. Whether suPAR is associated with COVID-19-related AKI is unknown. METHODS In a multinational observational study of adult patients hospitalized for COVID-19, we measured suPAR levels in plasma samples from 352 adult patients that had been collected within 48 hours of admission. We examined the association between suPAR levels and incident in-hospital AKI. RESULTS Of the 352 patients (57.4% were male, 13.9% were black, and mean age was 61 years), 91 (25.9%) developed AKI during their hospitalization, of whom 25 (27.4%) required dialysis. The median suPAR level was 5.61 ng/ml. AKI incidence rose with increasing suPAR tertiles, from a 6.0% incidence in patients with suPAR <4.60 ng/ml (first tertile) to a 45.8% incidence of AKI in patients with suPAR levels >6.86 ng/ml (third tertile). None of the patients with suPAR <4.60 ng/ml required dialysis during their hospitalization. In multivariable analysis, the highest suPAR tertile was associated with a 9.15-fold increase in the odds of AKI (95% confidence interval [95% CI], 3.64 to 22.93) and a 22.86-fold increase in the odds of requiring dialysis (95% CI, 2.77 to 188.75). The association was independent of inflammatory markers and persisted across subgroups. CONCLUSIONS Admission suPAR levels in patients hospitalized for COVID-19 are predictive of in-hospital AKI and the need for dialysis. SuPAR may be a key component of the pathophysiology of AKI in COVID-19.
Collapse
Affiliation(s)
- Tariq U Azam
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Husam R Shadid
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Patrick O'Hayer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Hanna Berlin
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Michael Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Peiyao Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Rodica Pop-Busui
- Division of Endocrinology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Izzet Altintas
- Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Jens Tingleff
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Marius A Stauning
- Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Maria-Evangelia Adami
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nicky Solomonidi
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Tsilika
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Pinkus Tober-Lau
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eleni Arnaoutoglou
- Department of Anesthesiology, School of Health Sciences, Faculty of Medicine, University of Thessaly, Thessaly, Greece
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Athanasios Chalkias
- Department of Anesthesiology, School of Health Sciences, Faculty of Medicine, University of Thessaly, Thessaly, Greece
| | - Sven H Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
46
|
Terpos E, Ntanasis-Stathopoulos I, Papassotiriou GP, Kastritis E, Margeli A, Kanellias N, Eleutherakis-Papaiakovou E, Migkou M, Fotiou D, Roussou M, Gavriatopoulou M, Malandrakis P, Psimenou E, Papassotiriou I, Dimopoulos MA. Circulating Soluble Urokinase-Type Plasminogen Activator Receptor Levels Reflect Renal Function in Newly Diagnosed Patients with Multiple Myeloma Treated with Bortezomib-Based Induction. J Clin Med 2020; 9:jcm9103201. [PMID: 33022958 PMCID: PMC7600599 DOI: 10.3390/jcm9103201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/26/2020] [Accepted: 10/02/2020] [Indexed: 12/04/2022] Open
Abstract
(1) Background: Soluble urokinase-type plasminogen activator receptor (suPAR) has been implicated in the pathogenesis of kidney disease in different disease settings. The aim of this study was to investigate a possible link between suPAR circulating levels and renal impairment (RI) in newly diagnosed patients with symptomatic multiple myeloma (NDMM) before and after frontline therapy with bortezomib-based regimens. (2) Methods: We studied 47 NDMM patients (57% males, median age 69.5 years) before the administration of anti-myeloma treatment and at best response to bortezomib-based therapy. suPAR was measured in the serum of all patients and of 24 healthy matched controls, using an immuno-enzymatic assay (ViroGates, Denmark). (3) Results: suPAR levels were elevated in NDMM patients at diagnosis compared to healthy individuals (p < 0.001). suPAR levels strongly correlated with disease stage (p-ANOVA < 0.001). suPAR levels both at diagnosis and at best response negatively correlated with estimated glomerular filtration rate (eGFR) values (p < 0.001). Interestingly, no significance changes in suPAR levels were observed at best response compared to baseline values (p = 0.31) among 18 responding patients with baseline eGFR < 50 mL/min/1.73 m2. (4) Conclusions: SuPAR levels reflect renal function in NDMM patients treated with bortezomib-based induction. Responders may have elevated circulating suPAR levels, possibly reflecting persistent kidney damage, despite their renal response.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
- Correspondence: ; Tel.: +30-213-2162846; Fax: +30-213-2162511
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Gerasimos-Petros Papassotiriou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Alexandra Margeli
- Department of Clinical Biochemistry, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (A.M.); (I.P.)
| | - Nikolaos Kanellias
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Evangelos Eleutherakis-Papaiakovou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Magdalini Migkou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Despina Fotiou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Maria Roussou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Erasmia Psimenou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece; (A.M.); (I.P.)
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.N.-S.); (G.-P.P.); (E.K.); (N.K.); (E.E.-P.); (M.M.); (D.F.); (M.R.); (M.G.); (P.M.); (E.P.); (M.A.D.)
| |
Collapse
|
47
|
Chalkias A, Mouzarou A, Samara E, Xanthos T, Ischaki E, Pantazopoulos I. Soluble Urokinase Plasminogen Activator Receptor: A Biomarker for Predicting Complications and Critical Care Admission of COVID-19 Patients. Mol Diagn Ther 2020; 24:517-521. [PMID: 32613288 PMCID: PMC7327460 DOI: 10.1007/s40291-020-00481-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The novel coronavirus infection has spread worldwide, causing a wide spectrum of clinical manifestations. Most patients develop moderate clinical illness, but a substantial number will experience severe pneumonia, which may rapidly progress to acute respiratory distress syndrome and multiple organ failure. In this population, soluble urokinase plasminogen activator receptor (suPAR) could serve as a quick triage test and independent marker of clinical severity, hospital and intensive care unit admission, complications, and mortality.
Collapse
Affiliation(s)
- Athanasios Chalkias
- Department of Anesthesiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece.
| | - Angeliki Mouzarou
- Department of Cardiology, General Hospital of Paphos, Paphos, Cyprus
| | - Evangelia Samara
- Department of Anesthesiology, Tzaneio General Hospital, Piraeus, Greece
| | | | - Eleni Ischaki
- First Department of Critical Care Medicine and Pulmonary Services, Evangelismos Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Pantazopoulos
- Department of Emergency Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| |
Collapse
|
48
|
Abstract
Podocytopathies are kidney diseases in which direct or indirect podocyte injury drives proteinuria or nephrotic syndrome. In children and young adults, genetic variants in >50 podocyte-expressed genes, syndromal non-podocyte-specific genes and phenocopies with other underlying genetic abnormalities cause podocytopathies associated with steroid-resistant nephrotic syndrome or severe proteinuria. A variety of genetic variants likely contribute to disease development. Among genes with non-Mendelian inheritance, variants in APOL1 have the largest effect size. In addition to genetic variants, environmental triggers such as immune-related, infection-related, toxic and haemodynamic factors and obesity are also important causes of podocyte injury and frequently combine to cause various degrees of proteinuria in children and adults. Typical manifestations on kidney biopsy are minimal change lesions and focal segmental glomerulosclerosis lesions. Standard treatment for primary podocytopathies manifesting with focal segmental glomerulosclerosis lesions includes glucocorticoids and other immunosuppressive drugs; individuals not responding with a resolution of proteinuria have a poor renal prognosis. Renin-angiotensin system antagonists help to control proteinuria and slow the progression of fibrosis. Symptomatic management may include the use of diuretics, statins, infection prophylaxis and anticoagulation. This Primer discusses a shift in paradigm from patient stratification based on kidney biopsy findings towards personalized management based on clinical, morphological and genetic data as well as pathophysiological understanding.
Collapse
|
49
|
Nikorowitsch J, Borchardt T, Appelbaum S, Ojeda F, Lackner KJ, Schnabel RB, Blankenberg S, Zeller T, Karakas M. Cardio-Renal Biomarker Soluble Urokinase-Type Plasminogen Activator Receptor Is Associated With Cardiovascular Death and Myocardial Infarction in Patients With Coronary Artery Disease Independent of Troponin, C-Reactive Protein, and Renal Function. J Am Heart Assoc 2020; 9:e015452. [PMID: 32299288 PMCID: PMC7428542 DOI: 10.1161/jaha.119.015452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Risk stratification among patients with coronary artery disease (CAD) is of considerable interest due to the potential to guide secondary preventive therapies. Thus, we evaluated the predictive value of soluble urokinase-type plasminogen activator receptor (suPAR) levels for cardiovascular mortality and nonfatal myocardial infarction in patients with CAD. Methods and Results Plasma levels of suPAR were measured in a cohort of 1703 patients with documented CAD as evidenced by coronary angiography-including 626 patients with acute coronary syndrome and 1077 patients with stable angina pectoris. Cardiovascular death and/or nonfatal myocardial infarction were defined as main outcome measures. During a median follow-up of 3.5 years, suPAR levels reliably predicted cardiovascular death or myocardial infarction in CAD, evidenced by survival curves stratified for tertiles of suPAR levels. In Cox regression analyses, the hazard ratio for the prediction of cardiovascular death and/or myocardial infarction was 2.19 (P<0.001) in the overall cohort and 2.56 in the acute coronary syndrome cohort (P<0.001). Even after adjustment for common cardiovascular risk factors, renal function and the biomarkers C-reactive protein, N-terminal pro-B-type natriuretic peptide and high-sensitivity troponin I suPAR still enabled a reliable prediction of cardiovascular death or myocardial infarction with a hazard ratio of 1.61 (P=0.022) in the overall cohort and 2.22 (P=0.005) in the acute coronary syndrome cohort. Conclusions SuPAR has a strong and independent prognostic value in secondary prevention settings, and thereby might represent a valuable biomarker for risk estimation in CAD.
Collapse
Affiliation(s)
- Julius Nikorowitsch
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
| | - Tim Borchardt
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
| | - Sebastian Appelbaum
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
| | - Francisco Ojeda
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
| | - Karl J. Lackner
- Department of Laboratory MedicineUniversity Medical CenterJohannes Gutenberg University MainzMainzGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein‐MainMainzGermany
| | - Renate B. Schnabel
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, KielHamburgGermany
| | - Stefan Blankenberg
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, KielHamburgGermany
| | - Tanja Zeller
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, KielHamburgGermany
| | - Mahir Karakas
- Clinic of CardiologyUniversity Heart and Vascular Center HamburgHamburgGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, KielHamburgGermany
| |
Collapse
|
50
|
Stewart BJ, Clatworthy MR. Applying single-cell technologies to clinical pathology: progress in nephropathology. J Pathol 2020; 250:693-704. [PMID: 32125696 PMCID: PMC8651001 DOI: 10.1002/path.5417] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 12/13/2022]
Abstract
Cells represent the basic building blocks of living organisms. Accurate characterisation of cellular phenotype, intercellular signalling networks, and the spatial organisation of cells within organs is crucial to deliver a better understanding of the processes underpinning physiology, and the perturbations that lead to disease. Single-cell methodologies have increased rapidly in scale and scope in recent years and are set to generate important insights into human disease. Here, we review current practices in nephropathology, which are dominated by relatively simple morphological descriptions of tissue biopsies based on their appearance using light microscopy. Bulk transcriptomics have more recently been used to explore glomerular and tubulointerstitial kidney disease, renal cancer, and the responses to injury and alloimmunity in kidney transplantation, generating novel disease insights and prognostic biomarkers. These studies set the stage for single-cell transcriptomic approaches that reveal cell-type-specific gene expression patterns in health and disease. These technologies allow genome-wide disease susceptibility genes to be interpreted with the knowledge of the specific cell populations within organs that express them, identifying candidate cell types for further study. Single-cell technologies are also moving beyond assaying individual cellular transcriptomes, to measuring the epigenetic landscape of single cells. Single-cell antigen-receptor gene sequencing also enables specific T- and B-cell clones to be tracked in different tissues and disease states. In the coming years these rich 'multi-omic' descriptions of kidney disease will enable histopathological descriptions to be comprehensively integrated with molecular phenotypes, enabling better disease classification and prognostication and the application of personalised treatment strategies. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Benjamin J Stewart
- Department of MedicineUniversity of CambridgeCambridgeUK
- Cellular GeneticsWellcome Sanger InstituteCambridgeUK
- Cambridge NIHR Biomedical Research CentreAddenbrooke's HospitalCambridgeUK
| | - Menna R Clatworthy
- Department of MedicineUniversity of CambridgeCambridgeUK
- Cellular GeneticsWellcome Sanger InstituteCambridgeUK
- Cambridge NIHR Biomedical Research CentreAddenbrooke's HospitalCambridgeUK
| |
Collapse
|