51
|
Braden GL, Landry DL. The Next Frontier: Biomarkers and Artificial Intelligence Predicting Cardiorenal Outcomes in Diabetic Kidney Disease. KIDNEY360 2022; 3:1480-1483. [PMID: 36245646 PMCID: PMC9528371 DOI: 10.34067/kid.0003322022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/01/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Gregory L. Braden
- Division of Nephrology, Baystate Medical Center, Springfield, Massachusetts
| | - Daniel L. Landry
- Division of Nephrology, Baystate Medical Center, Springfield, Massachusetts
| |
Collapse
|
52
|
Copur S, Tanriover C, Yavuz F, Soler MJ, Ortiz A, Covic A, Kanbay M. Novel strategies in nephrology: what to expect from the future? Clin Kidney J 2022; 16:230-244. [PMID: 36755838 PMCID: PMC9900595 DOI: 10.1093/ckj/sfac212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Indexed: 11/14/2022] Open
Abstract
Chronic kidney disease (CKD) will become the fifth global case of death by 2040. Its largest impact is on premature mortality but the number of persons with kidney failure requiring renal replacement therapy (RRT) is also increasing dramatically. Current RRT is suboptimal due to the shortage of kidney donors and dismal outcomes associated with both hemodialysis and peritoneal dialysis. Kidney care needs a revolution. In this review, we provide an update on emerging knowledge and technologies that will allow an earlier diagnosis of CKD, addressing the current so-called blind spot (e.g. imaging and biomarkers), and improve renal replacement therapies (wearable artificial kidneys, xenotransplantation, stem cell-derived therapies, bioengineered and bio-artificial kidneys).
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Cem Tanriover
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Furkan Yavuz
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Maria J Soler
- Department of Nephrology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Spain,Nephrology and Kidney Transplant Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Alberto Ortiz
- Department of Medicine, Universidad Autonoma de Madrid and IIS-Fundacion Jimenez Diaz, Madrid, Spain
| | - Adrian Covic
- Nephrology Clinic, Dialysis and Renal Transplant Center, ‘C.I. PARHON’ University Hospital, and ‘Grigore T. Popa’ University of Medicine, Iasi, Romania
| | | |
Collapse
|
53
|
Liu C, Debnath N, Mosoyan G, Chauhan K, Vasquez-Rios G, Soudant C, Menez S, Parikh CR, Coca SG. Systematic Review and Meta-Analysis of Plasma and Urine Biomarkers for CKD Outcomes. J Am Soc Nephrol 2022; 33:1657-1672. [PMID: 35858701 PMCID: PMC9529190 DOI: 10.1681/asn.2022010098] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 06/02/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Sensitive and specific biomarkers are needed to provide better biologic insight into the risk of incident and progressive CKD. However, studies have been limited by sample size and design heterogeneity. METHODS In this assessment of the prognostic value of preclinical plasma and urine biomarkers for CKD outcomes, we searched Embase (Ovid), MEDLINE ALL (Ovid), and Scopus up to November 30, 2020, for studies exploring the association between baseline kidney biomarkers and CKD outcomes (incident CKD, CKD progression, or incident ESKD). We used random-effects meta-analysis. RESULTS After screening 26,456 abstracts and 352 full-text articles, we included 129 studies in the meta-analysis for the most frequently studied plasma biomarkers (TNFR1, FGF23, TNFR2, KIM-1, suPAR, and others) and urine biomarkers (KIM-1, NGAL, and others). For the most frequently studied plasma biomarkers, pooled RRs for CKD outcomes were 2.17 (95% confidence interval [95% CI], 1.91 to 2.47) for TNFR1 (31 studies); 1.21 (95% CI, 1.15 to 1.28) for FGF-23 (30 studies); 2.07 (95% CI, 1.82 to 2.34) for TNFR2 (23 studies); 1.51 (95% CI, 1.38 to 1.66) for KIM-1 (18 studies); and 1.42 (95% CI, 1.30 to 1.55) for suPAR (12 studies). For the most frequently studied urine biomarkers, pooled RRs were 1.10 (95% CI, 1.05 to 1.16) for KIM-1 (19 studies) and 1.12 (95% CI, 1.06 to 1.19) for NGAL (19 studies). CONCLUSIONS Studies of preclinical biomarkers for CKD outcomes have considerable heterogeneity across study cohorts and designs, limiting comparisons of prognostic performance across studies. Plasma TNFR1, FGF23, TNFR2, KIM-1, and suPAR were among the most frequently investigated in the setting of CKD outcomes.
Collapse
Affiliation(s)
- Caroline Liu
- Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Neha Debnath
- Department of Medicine, Icahn School of Medicine at Mount Sinai (Morningside/West), New York, New York
| | - Gohar Mosoyan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kinsuk Chauhan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - George Vasquez-Rios
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Celine Soudant
- Division of Technology, Memorial Sloan Kettering Cancer Center Medical Library, New York, New York
| | - Steve Menez
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chirag R. Parikh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven G. Coca
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
54
|
Chang YK, Fan HC, Lin CC, Wang YH, Tsai WN, Lim PS. Association between atrial fibrillation and risk of end-stage renal disease among adults with diabetes mellitus. PLoS One 2022; 17:e0273646. [PMID: 36026496 PMCID: PMC9417190 DOI: 10.1371/journal.pone.0273646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/14/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes mellitus (DM) is an important risk factor in patients with end-stage renal disease (ESRD). DM is associated with the development of cardiovascular diseases, such as atrial fibrillation (AF), due to poor glycemic control. However, few studies have focused on the risk of developing ESRD among DM patients with and without AF. This study evaluated ESRD risk among DM patients with and without AF in Taiwan. Data were retrieved from one million patients randomly sampled from Taiwan’s National Health Insurance Research Database, including 6,105 DM patients with AF propensity score–matched with 6,105 DM patients without AF. Both groups were followed until death, any dialysis treatment, or December 31, 2013, whichever occurred first. AF was diagnosed by a qualified physician according to the International Classification of Diseases, 9th Revision, Clinical Modification (ICD-9-CM), using the diagnostic code 427.31. Patients aged <20 years or diagnosed with ESRD before the index date were excluded. A Cox proportional hazard regression model was used to calculate the relative ESRD risk. Among DM patients, those with AF have more comorbidities than those without AF. We also found a 1.18-fold (95% confidence interval [CI]: 1.01–1.46) increase in ESRD risk among patients with AF compared with those without AF. In addition, DM patients with hypertension, chronic kidney disease (CKD), or higher Charlson Comorbidity Index scores also have significantly increased ESRD risks than those without these complications. A 1.39-fold (95% CI: 1.04–1.86) increase in risk was observed for patients with AF among the non-CKD group. Our findings suggest that patients with DM should be closely monitored for irregular or rapid heart rates.
Collapse
Affiliation(s)
- Yu-Kang Chang
- Department of Medical Research, Tungs’ Taichung MetroHarbor Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Nursing, Jenteh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung MetroHarbor Hospital, Taichung, Taiwan
- Department of Rehabilitation, Jenteh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Chi-Chien Lin
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biomedical Science, iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
| | - Yuan-Hung Wang
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New City, Taiwan
| | - Wan-Ni Tsai
- Department of Endocrinology and Metabolism, Tungs’ Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Paik-Seong Lim
- Division of Renal Medicine, Tungs’ Taichung MetroHarbor Hospital, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
55
|
The unique second wave phenomenon in contrast enhanced ultrasound imaging with nanobubbles. Sci Rep 2022; 12:13619. [PMID: 35948582 PMCID: PMC9365822 DOI: 10.1038/s41598-022-17756-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/30/2022] [Indexed: 12/19/2022] Open
Abstract
Investigation of nanobubble (NB) pharmacokinetics in contrast-enhanced ultrasound (CEUS) at the pixel level shows a unique phenomenon where the first pass of the contrast agent bolus is accompanied by a second wave. This effect has not been previously observed in CEUS with microbubbles. The objective of this study was to investigate this second-wave phenomenon and its potential clinical applications. Seven mice with a total of fourteen subcutaneously-implanted tumors were included in the experiments. After injecting a bolus of NBs, the NB-CEUS images were acquired to record the time-intensity curves (TICs) at each pixel. These TICs are fitted to a pharmacokinetic model which we designed to describe the observed second-wave phenomenon. The estimated model parameters are presented as parametric maps to visualize the characteristics of tumor lesions. Histological analysis was also conducted in one mouse to compare the molecular features of tumor tissue with the obtained parametric maps. The second-wave phenomenon is evidently shown in a series of pixel-based TICs extracted from either tumor or tissues. The value of two model parameters, the ratio of the peak intensities of the second over the first wave, and the decay rate of the wash-out process present large differences between malignant tumor and normal tissue (0.04 < Jessen-Shannon divergence < 0.08). The occurrence of a second wave is a unique phenomenon that we have observed in NB-CEUS imaging of both mouse tumor and tissue. As the characteristics of the second wave are different between tumor and tissue, this phenomenon has the potential to support the diagnosis of cancerous lesions.
Collapse
|
56
|
Kobayashi H, Looker HC, Satake E, D’Addio F, Wilson JM, Saulnier PJ, Md Dom ZI, O’Neil K, Ihara K, Krolewski B, Badger HS, Petrazzuolo A, Corradi D, Galecki A, Wilson P, Najafian B, Mauer M, Niewczas MA, Doria A, Humphreys B, Duffin KL, Fiorina P, Nelson RG, Krolewski AS. Neuroblastoma suppressor of tumorigenicity 1 is a circulating protein associated with progression to end-stage kidney disease in diabetes. Sci Transl Med 2022; 14:eabj2109. [PMID: 35947673 PMCID: PMC9531292 DOI: 10.1126/scitranslmed.abj2109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Circulating proteins associated with transforming growth factor-β (TGF-β) signaling are implicated in the development of diabetic kidney disease (DKD). It remains to be comprehensively examined which of these proteins are involved in the pathogenesis of DKD and its progression to end-stage kidney disease (ESKD) in humans. Using the SOMAscan proteomic platform, we measured concentrations of 25 TGF-β signaling family proteins in four different cohorts composed in total of 754 Caucasian or Pima Indian individuals with type 1 or type 2 diabetes. Of these 25 circulating proteins, we identified neuroblastoma suppressor of tumorigenicity 1 (NBL1, aliases DAN and DAND1), a small secreted protein known to inhibit members of the bone morphogenic protein family, to be most strongly and independently associated with progression to ESKD during 10-year follow-up in all cohorts. The extent of damage to podocytes and other glomerular structures measured morphometrically in 105 research kidney biopsies correlated strongly with circulating NBL1 concentrations. Also, in vitro exposure to NBL1 induced apoptosis in podocytes. In conclusion, circulating NBL1 may be involved in the disease process underlying progression to ESKD, and its concentration in circulation may identify subjects with diabetes at increased risk of progression to ESKD.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Hypertension, and Endocrinology, Nihon University School of Medicine, Tokyo, Japan
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Eiichiro Satake
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Francesca D’Addio
- Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università di Milano and Endocrinology Division ASST Sacco-FBF, Milan, Italy
| | - Jonathan M. Wilson
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Pierre Jean. Saulnier
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
- CHU Poitiers, University of Poitiers, Inserm, Clinical Investigation Center CIC1402, Poitiers, France
| | - Zaipul I. Md Dom
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kristina O’Neil
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
| | - Katsuhito Ihara
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bozena Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hannah S. Badger
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Adriana Petrazzuolo
- Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università di Milano and Endocrinology Division ASST Sacco-FBF, Milan, Italy
| | - Domenico Corradi
- Department of Medicine and Surgery, Unit of Pathology, University of Parma, Parma, Italy
| | - Andrzej Galecki
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Parker Wilson
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University in Saint Louis School of Medicine, St. Louis, USA
| | - Behzad Najafian
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Michael Mauer
- Department of Pediatrics and Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Monika A. Niewczas
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Benjamin Humphreys
- Division of Nephrology, Department of Medicine, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Kevin L. Duffin
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Paolo Fiorina
- Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università di Milano and Endocrinology Division ASST Sacco-FBF, Milan, Italy
- Nephrology Division, Boston Children’s Hospital, Boston, MA, USA
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Andrzej S. Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
57
|
Rico-Fontalvo J, Aroca G, Cabrales J, Daza-Arnedo R, Yánez-Rodríguez T, Martínez-Ávila MC, Uparella-Gulfo I, Raad-Sarabia M. Molecular Mechanisms of Diabetic Kidney Disease. Int J Mol Sci 2022; 23:ijms23158668. [PMID: 35955802 PMCID: PMC9369345 DOI: 10.3390/ijms23158668] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/18/2022] Open
Abstract
The inflammatory component of diabetic kidney disease has become of great interest in recent years, with genetic and epigenetic variants playing a fundamental role in the initiation and progression of the disease. Cells of the innate immune system play a major role in the pathogenesis of diabetic kidney disease, with a lesser contribution from the adaptive immune cells. Other components such as the complement system also play a role, as well as specific cytokines and chemokines. The inflammatory component of diabetic kidney disease is of great interest and is an active research field, with the hope to find potential innovative therapeutic targets.
Collapse
Affiliation(s)
- Jorge Rico-Fontalvo
- Colombian Nephrology Association, Bogotá 110221, Colombia
- Management of Technologies and Innovation, Department of Engineering, Universidad Simón Bolivar, Cl. 58 #55-132, Barranquilla 080002, Colombia
| | - Gustavo Aroca
- Colombian Nephrology Association, Bogotá 110221, Colombia
- Faculty of Medicine, Universidad Simón Bolívar, Barranquilla 080002, Colombia
| | - Jose Cabrales
- Nephrology Fellow, Stanford University School of Medicine, Palo Alto, CA 94305, USA
- Correspondence:
| | | | | | | | | | | |
Collapse
|
58
|
Fusfeld L, Murphy JT, Yoon Y, Kam LY, Peters KE, Lin Tan P, Shanik M, Turchin A. Evaluation of the clinical utility of the PromarkerD in-vitro test in predicting diabetic kidney disease and rapid renal decline through a conjoint analysis. PLoS One 2022; 17:e0271740. [PMID: 35913946 PMCID: PMC9342737 DOI: 10.1371/journal.pone.0271740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 07/06/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Early identification of patients at risk of developing diabetic kidney disease or rapid renal decline is imperative for appropriate patient management, but traditional methods of predicting renal decline are limited. OBJECTIVE This study evaluated the impact of PromarkerD, a biomarker-based blood test predicting the risk of diabetic kidney disease (DKD) and rapid renal decline. METHODS Conjoint analysis clarified the importance of PromarkerD and other patient attributes to physician decisions for type 2 diabetes patients. Forty-two patient profiles were generated, with varying levels of albuminuria, estimated glomerular filtration rate (eGFR), blood pressure, hemoglobin A1c (HbA1c), age, and PromarkerD result. A web-based survey asked each physician to make monitoring/treatment decisions about eight randomly selected profiles. Data were analyzed using multivariable logit models. RESULTS Two hundred three primary care physicians and 197 endocrinologists completed the survey. PromarkerD result was most important for increasing the frequency of risk factor monitoring. PromarkerD was second to HbA1c in importance for deciding to prescribe sodium/glucose cotransporter-2 inhibitors (SGLT2s) with a DKD indication, second to blood pressure for increasing the dose of lisinopril, and second to eGFR for replacing ibuprofen with a non-nephrotoxic medication. Compared with no PromarkerD results, a high-risk PromarkerD result was associated with significantly higher odds of increasing monitoring frequency (odds ratio [OR]: 2.56, 95% confidence interval: 1.90-3.45), prescribing SGLT2s (OR: 1.98 [1.56-2.52]), increasing lisinopril dose (OR: 1.48 [1.17-1.87]), and replacing ibuprofen (OR: 1.78 [1.32-2.40]). A low-risk PromarkerD result was associated with significantly lower odds of increasing monitoring frequency (OR: 0.48 [0.37-0.64]), prescribing SGLT2s (OR: 0.70 [0.56-0.88]), and replacing ibuprofen (OR: 0.75 [0.57-0.99]). CONCLUSION PromarkerD could increase adoption of renoprotective interventions in patients at high risk for renal decline and lower the likelihood of aggressive treatment in those at low risk. Further studies are needed to assess patient outcomes with PromarkerD in real-world practice.
Collapse
Affiliation(s)
- Lauren Fusfeld
- Boston Healthcare Associates (now a Veranex Company), Boston, MA, United States of Ameria
| | - Jessica T. Murphy
- Boston Healthcare Associates (now a Veranex Company), Boston, MA, United States of Ameria
| | - YooJin Yoon
- Boston Healthcare Associates (now a Veranex Company), Boston, MA, United States of Ameria
| | | | | | | | - Michael Shanik
- Stony Brook University Medical Center, Stony Brook, NY, United States of Ameria
- Endocrine Associates of Long Island, PC, Smithtown, NY, United States of Ameria
| | | |
Collapse
|
59
|
Sarnak MJ, Katz R, Ix JH, Kimmel PL, Bonventre JV, Schelling J, Cushman M, Vasan RS, Waikar SS, Greenberg JH, Parikh CR, Coca SG, Sabbisetti V, Jogalekar MP, Rebholz C, Zheng Z, Gutierrez OM, Shlipak MG. Plasma Biomarkers as Risk Factors for Incident CKD. Kidney Int Rep 2022; 7:1493-1501. [PMID: 35812266 PMCID: PMC9263237 DOI: 10.1016/j.ekir.2022.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction Earlier identification of individuals at high risk of chronic kidney disease (CKD) may facilitate improved risk factor mitigation. Methods We evaluated the association of novel plasma biomarkers with incident CKD using a case-cohort design in participants without diabetes and with baseline estimated glomerular filtration rate (eGFR) ≥ 60 ml/min per 1.73 m2 in the Multi-Ethnic Study of Atherosclerosis (MESA) and Reasons for Geographic and Racial Differences in Stroke (REGARDS) cohorts. Incident CKD was defined as development of eGFR < 60 ml/min per 1.73 m2 and ≥40% decline in eGFR from baseline. We measured plasma markers of inflammation/fibrosis-soluble tumor necrosis factor receptors (TNFRs) 1 and 2 (TNFR-1 and TNFR-2), monocyte chemotactic protein-1 (MCP-1), chitinase 3-like protein 1 (YKL-40), and soluble urokinase-type plasminogen activator receptor (suPAR)-and tubular injury (kidney injury molecule 1 [KIM-1]). Cox regression models weighted for the case-cohort design were used to estimate hazard ratios (HRs) of incident CKD after adjustment for CKD risk factors, eGFR, and albuminuria. Results In MESA (median follow-up of 9.2 years), there were 497 individuals in the random subcohort and 163 incident CKD cases. In REGARDS (median follow-up of 9.4 years), there were 497 individuals in the random subcohort and 497 incident CKD cases. Each 2-fold higher plasma KIM-1 (adjusted HR 1.38 [95% CI 1.05-1.81]), suPAR (1.96 [1.10-3.49]), TNFR-1 (1.65 [1.04-2.62]), TNFR-2 (2.02 [1.21-3.38]), and YKL-40 (1.38 [1.09-1.75]) concentrations were associated with incident CKD in MESA. In REGARDS, TNFR-1 (1.99 [1.43-2.76]) and TNFR-2 (1.76 [1.22-2.54]) were associated with incident CKD. Conclusion Plasma concentrations of soluble TNFR-1 and TNFR-2 are consistently associated with incident CKD in nondiabetic community-living individuals in MESA and REGARDS.
Collapse
Affiliation(s)
- Mark J. Sarnak
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego School of Medicine, San Diego, California, USA
| | - Paul L. Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph V. Bonventre
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Mary Cushman
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, USA
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, USA
| | - Ramachandran S. Vasan
- Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Jason H. Greenberg
- Section of Nephrology, Department of Pediatrics, Program of Applied Translational Research, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chirag R. Parikh
- Section of Nephrology, Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Steven G. Coca
- Division of Nephrology, Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Venkata Sabbisetti
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manasi P. Jogalekar
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Casey Rebholz
- Department of Epidemiology and Statistics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Orlando M. Gutierrez
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael G. Shlipak
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Healthcare System, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
60
|
Gutiérrez OM, Shlipak MG, Katz R, Waikar SS, Greenberg JH, Schrauben SJ, Coca S, Parikh CR, Vasan RS, Feldman HI, Kimmel PL, Cushman M, Bonventre JV, Sarnak MJ, Ix JH. Associations of Plasma Biomarkers of Inflammation, Fibrosis, and Kidney Tubular Injury With Progression of Diabetic Kidney Disease: A Cohort Study. Am J Kidney Dis 2022; 79:849-857.e1. [PMID: 34752914 PMCID: PMC9072594 DOI: 10.1053/j.ajkd.2021.09.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/07/2021] [Indexed: 01/08/2023]
Abstract
RATIONALE & OBJECTIVE Most circulating biomarkers of chronic kidney disease (CKD) progression focus on factors reflecting glomerular filtration. Few biomarkers capture nonglomerular pathways of kidney injury or damage, which may be particularly informative in populations at high risk for CKD progression such as individuals with diabetes. STUDY DESIGN Cohort study. SETTING & PARTICIPANTS 594 participants (mean age, 70 years; 53% women) of the Reasons for Geographic and Racial Differences in Stroke (REGARDS) study who had diabetes and an estimated glomerular filtration rate (eGFR)<60mL/min/1.73m2 at baseline. EXPOSURES Plasma biomarkers of inflammation/fibrosis (TNFR1 and TNFR2, suPAR, MCP-1, YKL-40) and tubular injury (KIM-1) measured at the baseline visit. OUTCOMES Incident kidney failure with replacement therapy (KFRT). ANALYTICAL APPROACH Cox proportional hazards regression and least absolute shrinkage and selection operator regression adjusted for established risk factors for kidney function decline, baseline eGFR, and urinary albumin-creatinine ratio (UACR). RESULTS A total of 98 KFRT events were observed over a mean of 6.2±3.5 (standard deviation) years of follow-up. Plasma biomarkers were modestly associated with baseline eGFR (correlation coefficients ranging from-0.08 to-0.65) and UACR (0.14 to 0.56). In individual biomarker models adjusted for eGFR, UACR, and established risk factors, hazard ratios for incident KFRT per 2-fold higher biomarker concentrations were 1.52 (95% CI, 1.25-1.84) for plasma KIM-1, 1.54 (95% CI, 1.08-2.21) for TNFR1, 1.91 (95% CI, 1.16-3.14) for TNFR2, and 1.39 (95% CI, 1.05-1.84) for YKL-40. In least absolute shrinkage and selection operator regression models accounting for biomarkers in parallel, plasma KIM-1 and TNFR1 remained associated with incident KFRT. LIMITATIONS Single biomarker measurement, lack of follow-up eGFR assessments. CONCLUSIONS Individual plasma markers of inflammation/fibrosis (TNFR1, TNFR2, YKL-40) and tubular injury (KIM-1) were associated with risk of incident KFRT in adults with diabetes and an eGFR<60mL/min/1.73m2 after adjustment for established risk factors.
Collapse
Affiliation(s)
- Orlando M Gutiérrez
- Departments of Medicine and Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Michael G Shlipak
- Kidney Health Research Collaborative, Department of Medicine, San Francisco VA Healthcare System and University of California, San Francisco, San Francisco, California
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Jason H Greenberg
- Section of Nephrology, Department of Pediatrics, Program of Applied Translational Research, Yale University School of Medicine, New Haven, Connecticut
| | - Sarah J Schrauben
- Departments of Medicine and Biostatistics, Epidemiology and Informatics and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven Coca
- Division of Nephrology, Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Chirag R Parikh
- Section of Nephrology, Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ramachandran S Vasan
- Departments of Medicine and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Harold I Feldman
- Departments of Medicine and Biostatistics, Epidemiology and Informatics and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul L Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Mary Cushman
- Departments of Medicine and Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont
| | - Joseph V Bonventre
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark J Sarnak
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Joachim H Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California
| |
Collapse
|
61
|
Provenzano M, Maritati F, Abenavoli C, Bini C, Corradetti V, La Manna G, Comai G. Precision Nephrology in Patients with Diabetes and Chronic Kidney Disease. Int J Mol Sci 2022; 23:5719. [PMID: 35628528 PMCID: PMC9144494 DOI: 10.3390/ijms23105719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetes is the leading cause of kidney failure and specifically, diabetic kidney disease (DKD) occurs in up to 30% of all diabetic patients. Kidney disease attributed to diabetes is a major contributor to the global burden of the disease in terms of clinical and socio-economic impact, not only because of the risk of progression to End-Stage Kidney Disease (ESKD), but also because of the associated increase in cardiovascular (CV) risk. Despite the introduction of novel treatments that allow us to reduce the risk of future outcomes, a striking residual cardiorenal risk has been reported. This risk is explained by both the heterogeneity of DKD and the individual variability in response to nephroprotective treatments. Strategies that have been proposed to improve DKD patient care are to develop novel biomarkers that classify with greater accuracy patients with respect to their future risk (prognostic) and biomarkers that are able to predict the response to nephroprotective treatment (predictive). In this review, we summarize the principal prognostic biomarkers of type 1 and type 2 diabetes and the novel markers that help clinicians to individualize treatments and the basis of the characteristics that predict an optimal response.
Collapse
Affiliation(s)
- Michele Provenzano
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS—Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.M.); (C.A.); (C.B.); (V.C.); (G.C.)
| | | | | | | | | | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS—Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.M.); (C.A.); (C.B.); (V.C.); (G.C.)
| | | |
Collapse
|
62
|
Chen TK, Coca SG, Estrella MM, Appel LJ, Coresh J, Thiessen Philbrook H, Obeid W, Fried LF, Heerspink HJ, Ix JH, Shlipak MG, Kimmel PL, Parikh CR, Grams ME. Longitudinal TNFR1 and TNFR2 and Kidney Outcomes: Results from AASK and VA NEPHRON-D. J Am Soc Nephrol 2022; 33:996-1010. [PMID: 35314457 PMCID: PMC9063900 DOI: 10.1681/asn.2021060735] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 02/23/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Higher baseline levels of soluble TNF receptors (TNFR1 and TNFR2) have been associated with progressive CKD. Whether longitudinal changes in these biomarkers of inflammation are also associated with worse kidney outcomes has been less studied. METHODS We evaluated associations of longitudinal changes in TNFR1 and TNFR2 with ESKD in the African American Study of Kidney Disease and Hypertension (AASK; 38% female; 0% diabetes) and kidney function decline (first occurrence of ≥30 ml/min per 1.73 m2 or ≥50% eGFR decline if randomization eGFR ≥60 or <60 ml/min per 1.73 m2, respectively; ESKD) in the Veterans Affairs Nephropathy in Diabetes trial (VA NEPHRON-D; 99% male; 100% diabetes) using Cox models. Biomarkers were measured from samples collected at 0-, 12-, and 24-month visits for AASK (serum) and 0- and 12-month visits for VA NEPHRON-D (plasma). Biomarker slopes (AASK) were estimated using linear mixed-effects models. Covariates included sociodemographic/clinical factors, baseline biomarker level, and kidney function. RESULTS There were 129 ESKD events over a median of 7.0 years in AASK (n=418) and 118 kidney function decline events over a median of 1.5 years in VA NEPHRON-D (n=754). In AASK, each 1 SD increase in TNFR1 and TNFR2 slope was associated with 2.98- and 1.87-fold higher risks of ESKD, respectively. In VA NEPHRON-D, each 1 SD increase in TNFR1 and TNFR2 was associated with 3.20- and 1.43-fold higher risks of kidney function decline, respectively. CONCLUSIONS Among individuals with and without diabetes, longitudinal increases in TNFR1 and TNFR2 were each associated with progressive CKD, independent of initial biomarker level and kidney function.
Collapse
Affiliation(s)
- Teresa K. Chen
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Steven G. Coca
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michelle M. Estrella
- Kidney Health Research Collaborative and Division of Nephrology, Department of Medicine, University of California and San Francisco VA Health Care System, San Francisco, California
| | - Lawrence J. Appel
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Josef Coresh
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Wassim Obeid
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Linda F. Fried
- Renal Section, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- Departments of Medicine, Epidemiology, and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, and Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Michael G. Shlipak
- Kidney Health Research Collaborative and Division of Nephrology, Department of Medicine, University of California and San Francisco VA Health Care System, San Francisco, California
| | - Paul L. Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Morgan E. Grams
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
63
|
Inflammatory biomarkers in staging of chronic kidney disease: elevated TNFR2 levels accompanies renal function decline. Inflamm Res 2022; 71:591-602. [PMID: 35471601 DOI: 10.1007/s00011-022-01574-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Inflammation is a common feature in the pathogenesis of chronic kidney disease (CKD), regardless of the disease cause. Our aim was to evaluate the potential of several inflammatory biomarkers in CKD diagnosis and staging. METHODS A total of 24 healthy controls and 92 pre-dialysis CKD patients with diverse etiologies, were enrolled in this study and grouped according to their CKD stage. We analysed the circulating levels of inflammatory molecules, C-reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), tumor necrosis factor receptor 2 (TNFR2), pentraxin 3 (PTX3) and leptin, as well as the hemogram. We studied their association with parameters of kidney function and kidney injury, to evaluate their potential as early markers of the disease and/or of its worsening, as well as their interplay. RESULTS Compared to controls, patients in CKD stages 1-2 presented significantly higher IL-6 and TNFR2 levels, and higher neutrophil-to-lymphocyte ratio. All inflammatory cytokines and acute-phase proteins showed a trend to increase up to stage 3, stabilizing or declining thereafter, save for TNFR2, which steadily increased from stage to stage. All inflammatory molecules, apart from PTX3, were negatively and significantly correlated with eGFR, with a remarkable value for TNFR2 (r = - 0.732, p < 0.001). CONCLUSION TNFR2 might be useful for an early detection of CKD, as well as for disease staging/worsening. Still, the potential value of this biomarker in disease progression warrants further investigation.
Collapse
|
64
|
Chan KW, Yu KY, Yiu WH, Xue R, Lok SWY, Li H, Zou Y, Ma J, Lai KN, Tang SCW. Potential Therapeutic Targets of Rehmannia Formulations on Diabetic Nephropathy: A Comparative Network Pharmacology Analysis. Front Pharmacol 2022; 13:794139. [PMID: 35387335 PMCID: PMC8977554 DOI: 10.3389/fphar.2022.794139] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/10/2022] [Indexed: 11/14/2022] Open
Abstract
Background: Previous retrospective cohorts showed that Rehmannia-6 (R-6, Liu-wei-di-huang-wan) formulations were associated with significant kidney function preservation and mortality reduction among chronic kidney disease patients with diabetes. This study aimed to investigate the potential mechanism of action of common R-6 variations in a clinical protocol for diabetic nephropathy (DN) from a system pharmacology approach. Study Design and Methods: Disease-related genes were retrieved from GeneCards and OMIM by searching “Diabetic Nephropathy” and “Macroalbuminuria”. Variations of R-6 were identified from a published existing clinical practice guideline developed from expert consensus and pilot clinical service program. The chemical compound IDs of each herb were retrieved from TCM-Mesh and PubChem. Drug targets were subsequently revealed via PharmaMapper and UniProtKB. The disease gene interactions were assessed through STRING, and disease–drug protein–protein interaction network was integrated and visualized by Cytoscape. Clusters of disease–drug protein–protein interaction were constructed by Molecular Complex Detection (MCODE) extension. Functional annotation of clusters was analyzed by DAVID and KEGG pathway enrichment. Differences among variations of R-6 were compared. Binding was verified by molecular docking with AutoDock. Results: Three hundred fifty-eight genes related to DN were identified, forming 11 clusters which corresponded to complement and coagulation cascades and signaling pathways of adipocytokine, TNF, HIF-1, and AMPK. Five variations of R-6 were analyzed. Common putative targets of the R-6 variations on DN included ACE, APOE, CCL2, CRP, EDN1, FN1, HGF, ICAM1, IL10, IL1B, IL6, INS, LEP, MMP9, PTGS2, SERPINE1, and TNF, which are related to regulation of nitric oxide biosynthesis, lipid storage, cellular response to lipopolysaccharide, inflammatory response, NF-kappa B transcription factor activity, smooth muscle cell proliferation, blood pressure, cellular response to interleukin-1, angiogenesis, cell proliferation, peptidyl-tyrosine phosphorylation, and protein kinase B signaling. TNF was identified as the seed for the most significant cluster of all R-6 variations. Targets specific to each formulation were identified. The key chemical compounds of R-6 have good binding ability to the putative protein targets. Conclusion: The mechanism of action of R-6 on DN is mostly related to the TNF signaling pathway as a core mechanism, involving amelioration of angiogenesis, fibrosis, inflammation, disease susceptibility, and oxidative stress. The putative targets identified could be validated through clinical trials.
Collapse
Affiliation(s)
- Kam Wa Chan
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kam Yan Yu
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wai Han Yiu
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Rui Xue
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sarah Wing-Yan Lok
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hongyu Li
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yixin Zou
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jinyuan Ma
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kar Neng Lai
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | |
Collapse
|
65
|
Lousa I, Reis F, Santos-Silva A, Belo L. The Signaling Pathway of TNF Receptors: Linking Animal Models of Renal Disease to Human CKD. Int J Mol Sci 2022; 23:3284. [PMID: 35328704 PMCID: PMC8950598 DOI: 10.3390/ijms23063284] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic kidney disease (CKD) has been recognized as a global public health problem. Despite the current advances in medicine, CKD-associated morbidity and mortality remain unacceptably high. Several studies have highlighted the contribution of inflammation and inflammatory mediators to the development and/or progression of CKD, such as tumor necrosis factor (TNF)-related biomarkers. The inflammation pathway driven by TNF-α, through TNF receptors 1 (TNFR1) and 2 (TNFR2), involves important mediators in the pathogenesis of CKD. Circulating levels of TNFRs were associated with changes in other biomarkers of kidney function and injury, and were described as predictors of disease progression, cardiovascular morbidity, and mortality in several cohorts of patients. Experimental studies describe the possible downstream signaling pathways induced upon TNFR activation and the resulting biological responses. This review will focus on the available data on TNFR1 and TNFR2, and illustrates their contributions to the pathophysiology of kidney diseases, their cellular and molecular roles, as well as their potential as CKD biomarkers. The emerging evidence shows that TNF receptors could act as biomarkers of renal damage and as mediators of the disease. Furthermore, it has been suggested that these biomarkers could significantly improve the discrimination of clinical CKD prognostic models.
Collapse
Affiliation(s)
- Irina Lousa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (I.L.); (A.S.-S.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075 Coimbra, Portugal
| | - Alice Santos-Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (I.L.); (A.S.-S.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Luís Belo
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (I.L.); (A.S.-S.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
66
|
Jung CY, Yoo TH. Pathophysiologic Mechanisms and Potential Biomarkers in Diabetic Kidney Disease. Diabetes Metab J 2022; 46:181-197. [PMID: 35385633 PMCID: PMC8987689 DOI: 10.4093/dmj.2021.0329] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
Although diabetic kidney disease (DKD) remains the leading cause of end-stage kidney disease eventually requiring chronic kidney replacement therapy, the prevalence of DKD has failed to decline over the past 30 years. In order to reduce disease prevalence, extensive research has been ongoing to improve prediction of DKD onset and progression. Although the most commonly used markers of DKD are albuminuria and estimated glomerular filtration rate, their limitations have encouraged researchers to search for novel biomarkers that could improve risk stratification. Considering that DKD is a complex disease process that involves several pathophysiologic mechanisms such as hyperglycemia induced inflammation, oxidative stress, tubular damage, eventually leading to kidney damage and fibrosis, many novel biomarkers that capture one specific mechanism of the disease have been developed. Moreover, the increasing use of high-throughput omic approaches to analyze biological samples that include proteomics, metabolomics, and transcriptomics has emerged as a strong tool in biomarker discovery. This review will first describe recent advances in the understanding of the pathophysiology of DKD, and second, describe the current clinical biomarkers for DKD, as well as the current status of multiple potential novel biomarkers with respect to protein biomarkers, proteomics, metabolomics, and transcriptomics.
Collapse
Affiliation(s)
- Chan-Young Jung
- Department of Internal Medicine and Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine and Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea
- Corresponding author: Tae-Hyun Yoo https://orcid.org/0000-0002-9183-4507 Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea E-mail:
| |
Collapse
|
67
|
Kefaloyianni E. Soluble forms of cytokine and growth factor receptors: Mechanisms of generation and modes of action in the regulation of local and systemic inflammation. FEBS Lett 2022; 596:589-606. [PMID: 35113454 DOI: 10.1002/1873-3468.14305] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/16/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022]
Abstract
Cytokine and growth factor receptors are usually transmembrane proteins but they can also exist in soluble forms, either through cleavage and release of their ligand-binding extracellular domain, or through secretion of a soluble isoform. As an extension of this concept, transmembrane receptors on exosomes released into the circulation may act similarly to circulating soluble receptors. These soluble receptors add to the complexity of cytokine and growth factor signalling: they can function as decoy receptor that compete for ligand binding with their respective membrane-bound forms thereby attenuating signalling, or stabilize their ligands, or activate additional signalling events through interactions with other cell-surface proteins. Their soluble nature allows for a functional role away from the production sites, in remote cell types and organs. Accumulating evidence demonstrates that soluble receptors participate in the regulation and orchestration of various key cellular processes, particularly inflammatory responses. In this review, we will discuss release mechanisms of soluble cytokine and growth factor receptors, their mechanisms of action, as well as strategies for targeting their pathways in disease.
Collapse
Affiliation(s)
- Eirini Kefaloyianni
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
68
|
Waijer SW, Sen T, Arnott C, Neal B, Kosterink JG, Mahaffey KW, Parikh CR, de Zeeuw D, Perkovic V, Neuen BL, Coca SG, Hansen MK, Gansevoort RT, Heerspink HJ. Association between TNF Receptors and KIM-1 with Kidney Outcomes in Early-Stage Diabetic Kidney Disease. Clin J Am Soc Nephrol 2022; 17:251-259. [PMID: 34876454 PMCID: PMC8823939 DOI: 10.2215/cjn.08780621] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/29/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Clinical trials in nephrology are enriched for patients with micro- or macroalbuminuria to enroll patients at risk of kidney failure. However, patients with normoalbuminuria can also progress to kidney failure. TNF receptor-1, TNF receptor-2, and kidney injury marker-1 (KIM-1) are known to be associated with kidney disease progression in patients with micro- or macroalbuminuria. We assessed the value of TNF receptor-1, TNF receptor-2, and KIM-1 as prognostic biomarkers for CKD progression in patients with type 2 diabetes and normoalbuminuria. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS TNF receptor-1, TNF receptor-2, and KIM-1 were measured using immunoassays in plasma samples from patients with type 2 diabetes at high cardiovascular risk participating in the Canagliflozin Cardiovascular Assessment Study trial. We used multivariable adjusted Cox proportional hazards analyses to estimate hazard ratios per doubling of each biomarker for the kidney outcome, stratified the population by the fourth quartile of each biomarker distribution, and assessed the number of events and event rates. RESULTS In patients with normoalbuminuria (n=2553), 51 kidney outcomes were recorded during a median follow-up of 6.1 (interquartile range, 5.8-6.4) years (event rate, 3.5; 95% confidence interval, 2.6 to 4.6 per 1000 patient-years). Each doubling of baseline TNF receptor-1 (hazard ratio, 4.2; 95% confidence interval, 1.8 to 9.6) and TNF receptor-2 (hazard ratio, 2.3; 95% confidence interval, 1.5 to 3.6) was associated with a higher risk for the kidney outcome. Baseline KIM-1, urinary albumin-creatinine ratio, and eGFR were not associated with kidney outcomes. The event rates in the highest quartile of TNF receptor-1 (≥2992 ng/ml) and TNF receptor-2 (≥11,394 ng/ml) were 5.6 and 7.0 events per 1000 patient-years, respectively, compared with 2.8 and 2.3, respectively, in the lower three quartiles. CONCLUSIONS TNF receptor-1 and TNF receptor-2 are associated with kidney outcomes in patients with type 2 diabetes and normoalbuminuria. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER CANagliflozin cardioVascular Assessment Study (CANVAS), NCT01032629.
Collapse
Affiliation(s)
- Simke W. Waijer
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Taha Sen
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Clare Arnott
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Bruce Neal
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Jos G.W. Kosterink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands,Department of PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands
| | - Kenneth W. Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Chirag R. Parikh
- Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Dick de Zeeuw
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vlado Perkovic
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Brendon L. Neuen
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Steven G. Coca
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Ron T. Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hiddo J.L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands,The George Institute for Global Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
69
|
Kong L, Andrikopoulos S, MacIsaac RJ, Mackay LK, Nikolic‐Paterson DJ, Torkamani N, Zafari N, Marin ECS, Ekinci EI. Role of the adaptive immune system in diabetic kidney disease. J Diabetes Investig 2022; 13:213-226. [PMID: 34845863 PMCID: PMC8847140 DOI: 10.1111/jdi.13725] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/19/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic kidney disease (DKD) is a highly prevalent complication of diabetes and the leading cause of end-stage kidney disease. Inflammation is recognized as an important driver of progression of DKD. Activation of the immune response promotes a pro-inflammatory milieu and subsequently renal fibrosis, and a progressive loss of renal function. Although the role of the innate immune system in diabetic renal disease has been well characterized, the potential contribution of the adaptive immune system remains poorly defined. Emerging evidence in experimental models of DKD indicates an increase in the number of T cells in the circulation and in the kidney cortex, that in turn triggers secretion of inflammatory mediators such as interferon-γ and tumor necrosis factor-α, and activation of cells in innate immune response. In human studies, the number of T cells residing in the interstitial region of the kidney correlates with the degree of albuminuria in people with type 2 diabetes. Here, we review the role of the adaptive immune system, and associated cytokines, in the development of DKD. Furthermore, the potential therapeutic benefits of targeting the adaptive immune system as a means of preventing the progression of DKD are discussed.
Collapse
Affiliation(s)
- Lingyun Kong
- Department of MedicineAustin Health, University of MelbourneMelbourneVictoriaAustralia
| | | | - Richard J MacIsaac
- Department of MedicineAustin Health, University of MelbourneMelbourneVictoriaAustralia
- Department of Endocrinology & DiabetesSt Vincent's Hospital MelbourneMelbourneVictoriaAustralia
| | - Laura K Mackay
- Department of Microbiology and ImmunologyPeter Doherty Institute for Infection and ImmunityThe University of MelbourneMelbourneVictoriaAustralia
| | - David J Nikolic‐Paterson
- Department of NephrologyMonash Medical Center and Monash University Center for Inflammatory DiseasesMelbourneVictoriaAustralia
| | - Niloufar Torkamani
- Department of MedicineAustin Health, University of MelbourneMelbourneVictoriaAustralia
- Endocrine Center of ExcellenceAustin HealthMelbourneVictoriaAustralia
| | - Neda Zafari
- Department of MedicineAustin Health, University of MelbourneMelbourneVictoriaAustralia
| | - Evelyn C S Marin
- College of Sport and Exercise ScienceVictoria UniversityMelbourneVictoriaAustralia
| | - Elif I Ekinci
- Department of MedicineAustin Health, University of MelbourneMelbourneVictoriaAustralia
- Endocrine Center of ExcellenceAustin HealthMelbourneVictoriaAustralia
| |
Collapse
|
70
|
Guarneri M, Scola L, Giarratana RM, Bova M, Carollo C, Vaccarino L, Calandra L, Lio D, Balistreri CR, Cottone S. MIF rs755622 and IL6 rs1800795 Are Implied in Genetic Susceptibility to End-Stage Renal Disease (ESRD). Genes (Basel) 2022; 13:226. [PMID: 35205271 PMCID: PMC8872268 DOI: 10.3390/genes13020226] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic kidney disease (CKD) is characterized by an increased risk of kidney failure and end-stage renal disease (ESRD). Aging and comorbidities as cardiovascular diseases, metabolic disorders, infectious diseases, or tumors, might increase the risk of dialysis. In addition, genetic susceptibility factors might modulate kidney damage evolution. We have analyzed, in a group of ESRD patients and matched controls, a set of SNPs of genes (Klotho rs577912, rs564481, rs9536314; FGF23 rs7955866; IGF1 rs35767; TNFA rs1800629; IL6 rs1800795; MIF rs755622, rs1007888) chosen in relation to their possible involvement with renal disease and concomitant pathologies. Analysis of the raw data did indicate that IL6 rs180795 and MIF rs755622 SNPs might be markers of genetic susceptibility to ESRD. In particular, the C positive genotypes of MIF rs755622, (dominant model) seem to be an independent risk factor for ESDR patients (data adjusted for age, gender, and associated pathologies). Stratifying results according to age MIF rs755622 C positive genotype frequencies are increased in both the two age classes considered (<59 and ≥59-year-old subjects). Analyses of data according to gender allowed us to observe that ESRD women shoved a significantly reduced frequency of genotypes bearing IL6 rs180795 C allele. In addition, MIF rs755622 might interact with diabetes or hypercholesterolemia in increasing susceptibility to ESRD. In conclusion, our data indicate that some polymorphisms involved in the regulation of both renal function and inflammatory response can influence the evolution of chronic kidney disease and suggest that the modulation of the activities of these and other genes should also be considered as therapeutic targets on to intervene with innovative therapies.
Collapse
Affiliation(s)
- Marco Guarneri
- Unit of Nephrology & Hypertension, European Society of Hypertension Excellence Center, Department of Health Promotion Sciences, Maternal & Infant Care, Internal Medicine & Medical Specialties (PROMISE), University of Palermo, “Paolo Giaccone” University Hospital, 90127 Palermo, Italy; (M.G.); (C.C.); (L.C.); (S.C.)
| | - Letizia Scola
- Clinical Pathology, Department of Bio-Medicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy; (L.S.); (R.M.G.); (M.B.); (L.V.); (C.R.B.)
| | - Rosa Maria Giarratana
- Clinical Pathology, Department of Bio-Medicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy; (L.S.); (R.M.G.); (M.B.); (L.V.); (C.R.B.)
| | - Manuela Bova
- Clinical Pathology, Department of Bio-Medicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy; (L.S.); (R.M.G.); (M.B.); (L.V.); (C.R.B.)
| | - Caterina Carollo
- Unit of Nephrology & Hypertension, European Society of Hypertension Excellence Center, Department of Health Promotion Sciences, Maternal & Infant Care, Internal Medicine & Medical Specialties (PROMISE), University of Palermo, “Paolo Giaccone” University Hospital, 90127 Palermo, Italy; (M.G.); (C.C.); (L.C.); (S.C.)
| | - Loredana Vaccarino
- Clinical Pathology, Department of Bio-Medicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy; (L.S.); (R.M.G.); (M.B.); (L.V.); (C.R.B.)
| | - Leonardo Calandra
- Unit of Nephrology & Hypertension, European Society of Hypertension Excellence Center, Department of Health Promotion Sciences, Maternal & Infant Care, Internal Medicine & Medical Specialties (PROMISE), University of Palermo, “Paolo Giaccone” University Hospital, 90127 Palermo, Italy; (M.G.); (C.C.); (L.C.); (S.C.)
| | - Domenico Lio
- Clinical Pathology, Department of Bio-Medicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy; (L.S.); (R.M.G.); (M.B.); (L.V.); (C.R.B.)
| | - Carmela Rita Balistreri
- Clinical Pathology, Department of Bio-Medicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy; (L.S.); (R.M.G.); (M.B.); (L.V.); (C.R.B.)
| | - Santina Cottone
- Unit of Nephrology & Hypertension, European Society of Hypertension Excellence Center, Department of Health Promotion Sciences, Maternal & Infant Care, Internal Medicine & Medical Specialties (PROMISE), University of Palermo, “Paolo Giaccone” University Hospital, 90127 Palermo, Italy; (M.G.); (C.C.); (L.C.); (S.C.)
| |
Collapse
|
71
|
Peng L, Chen Y, Shi S, Wen H. Stem cell-derived and circulating exosomal microRNAs as new potential tools for diabetic nephropathy management. Stem Cell Res Ther 2022; 13:25. [PMID: 35073973 PMCID: PMC8785577 DOI: 10.1186/s13287-021-02696-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Despite major advances in the treatment of diabetic nephropathy (DN) in recent years, it remains the most common cause of end-stage renal disease. An early diagnosis and therapy may slow down the DN progression. Numerous potential biomarkers are currently being researched. Circulating levels of the kidney-released exosomes and biological molecules, which reflect the DN pathology including glomerular and tubular dysfunction as well as mesangial expansion and fibrosis, have shown the potential for predicting the occurrence and progression of DN. Moreover, many experimental therapies are currently being investigated, including stem cell therapy and medications targeting inflammatory, oxidant, or pro-fibrotic pathways activated during the DN progression. The therapeutic potential of stem cells is partly depending on their secretory capacity, particularly exosomal microRNAs (Exo-miRs). In recent years, a growing line of research has shown the participation of Exo-miRs in the pathophysiological processes of DN, which may provide effective therapeutic and biomarker tools for DN treatment. METHODS A systematic literature search was performed in MEDLINE, Scopus, and Google Scholar to collect published findings regarding therapeutic stem cell-derived Exo-miRs for DN treatment as well as circulating Exo-miRs as potential DN-associated biomarkers. FINDINGS Glomerular mesangial cells and podocytes are the most important culprits in the pathogenesis of DN and, thus, can be considered valuable therapeutic targets. Preclinical investigations have shown that stem cell-derived exosomes can exert beneficial effects in DN by transferring renoprotective miRs to the injured mesangial cells and podocytes. Of note, renoprotective Exo-miR-125a secreted by adipose-derived mesenchymal stem cells can improve the injured mesangial cells, while renoprotective Exo-miRs secreted by adipose-derived stem cells (Exo-miR-486 and Exo-miR-215-5p), human urine-derived stem cells (Exo-miR-16-5p), and bone marrow-derived mesenchymal stem cells (Exo-miR-let-7a) can improve the injured podocytes. On the other hand, clinical investigations have indicated that circulating Exo-miRs isolated from urine or serum hold great potential as promising biomarkers in DN.
Collapse
Affiliation(s)
- Lei Peng
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Yu Chen
- Department of Cardiology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Shaoqing Shi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Heling Wen
- Department of Cardiology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, 610072, China.
| |
Collapse
|
72
|
Da Silva Lodge M, Pullen N, Pereira M, Johnson TS. Urinary levels of pro-fibrotic transglutaminase 2 (TG2) may help predict progression of chronic kidney disease. PLoS One 2022; 17:e0262104. [PMID: 35041708 PMCID: PMC8765645 DOI: 10.1371/journal.pone.0262104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/16/2021] [Indexed: 01/04/2023] Open
Abstract
Renal clinical chemistry only detects kidney dysfunction after considerable damage has occurred and is imperfect in predicting long term outcomes. Consequently, more sensitive markers of early damage and better predictors of progression are being urgently sought, to better support clinical decisions and support shorter clinical trials. Transglutaminase 2 (TG2) is strongly implicated in the fibrotic remodeling that drives chronic kidney disease (CKD). We hypothesized that urinary TG2 and its ε-(γ-glutamyl)-lysine crosslink product could be useful biomarkers of kidney fibrosis and progression. Animal models: a rat 4-month 5/6th subtotal nephrectomy model of CKD and a rat 8-month streptozotocin model of diabetic kidney disease had 24-hour collection of urine, made using a metabolic cage, at regular periods throughout disease development. Patients: Urine samples from patients with CKD (n = 290) and healthy volunteers (n = 33) were collected prospectively, and progression tracked for 3 years. An estimated glomerular filtration rate (eGFR) loss of 2-5 mL/min/year was considered progressive, with rapid progression defined as > 5 mL/min/year. Assays: TG2 was measured in human and rat urine samples by enzyme-linked immunosorbent assay (ELISA) and ε-(γ-glutamyl)-lysine by exhaustive proteolytic digestion and amino acid analysis. Urinary TG2 and ε-(γ-glutamyl)-lysine increased with the development of fibrosis in both animal model systems. Urinary TG2 was 41-fold higher in patients with CKD than HVs, with levels elevated 17-fold by CKD stage 2. The urinary TG2:creatinine ratio (UTCR) was 9 ng/mmol in HV compared with 114 ng/mmol in non-progressive CKD, 1244 ng/mmol in progressive CKD and 1898 ng/mmol in rapidly progressive CKD. Both urinary TG2 and ε-(γ-glutamyl)-lysine were significantly associated with speed of progression in univariate logistic regression models. In a multivariate model adjusted for urinary TG2, ε-(γ-glutamyl)-lysine, age, sex, urinary albumin:creatinine ratio (UACR), urinary protein:creatinine ratio (UPCR), and CKD stage, only TG2 remained statistically significant. Receiver operating characteristic (ROC) curve analysis determined an 86.4% accuracy of prediction of progression for UTCR compared with 73.5% for UACR. Urinary TG2 and ε-(γ-glutamyl)-lysine are increased in CKD. In this pilot investigation, UTCR was a better predictor of progression in patients with CKD than UACR. Larger studies are now warranted to fully evaluate UTCR value in predicting patient outcomes.
Collapse
Affiliation(s)
- Michelle Da Silva Lodge
- Academic Nephrology Unit and Sheffield Kidney Institute, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Nick Pullen
- Pfizer Global Research and Development, Cambridge, MA, United States of America
| | - Miguel Pereira
- Statistical Sciences and Innovation, UCB Pharma, Slough, United Kingdom
| | - Timothy S. Johnson
- Academic Nephrology Unit and Sheffield Kidney Institute, University of Sheffield Medical School, Sheffield, United Kingdom
| |
Collapse
|
73
|
Murakoshi M, Gohda T, Sakuma H, Shibata T, Adachi E, Kishida C, Ichikawa S, Koshida T, Kamei N, Suzuki Y. Progranulin and Its Receptor Predict Kidney Function Decline in Patients With Type 2 Diabetes. Front Endocrinol (Lausanne) 2022; 13:849457. [PMID: 35432201 PMCID: PMC9012489 DOI: 10.3389/fendo.2022.849457] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Progranulin (PGRN), a growth factor, is abundantly expressed in a broad range of tissues and cell types with pleiotropic functions including inflammation, neurodegeneration, and facilitating lysosome acidification. PGRN binds to TNF receptors (TNFR) and inhibits downstream inflammatory signaling pathways. TNFR is a well-known predictor of glomerular filtration rate (GFR) decline in a variety of diseases. Therefore, we measured circulating PGRN in addition to TNFR using an enzyme-linked immunosorbent assay and explored whether it predicted renal prognosis in 201 Japanese patients with type 2 diabetes. During a median follow-up of 7.6 years, 21 participants reached primary renal endpoint, which involves a decline of at least 57% in eGFR from baseline, or the onset of end-stage renal disease. Univariate Cox regression analysis revealed that classical renal measures (GFR and albuminuria), two TNF-related biomarkers (PGRN and TNFR), and BMI were associated with this outcome. Multivariate analysis demonstrated that high levels of PGRN [HR 2.50 (95%CI 2.47-2.52)] or TNFR1 [HR 5.38 (95%CI 5.26-5.50)] were associated with this outcome after adjusting for relevant covariates. The high levels of PGRN as well as TNFR1 were associated with a risk of primary renal outcome in patients with type 2 diabetes after adjusting for established risk factors.
Collapse
Affiliation(s)
- Maki Murakoshi
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Tomohito Gohda
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
- *Correspondence: Tomohito Gohda,
| | - Hiroko Sakuma
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Terumi Shibata
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Eri Adachi
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Chiaki Kishida
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Saki Ichikawa
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Takeo Koshida
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Nozomu Kamei
- Department of Endocrinology and Metabolism, Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
74
|
Tokita J, Vega A, Sinfield C, Naik N, Rathi S, Martin S, Wang S, Amoruso L, Zabetian A, Coca SG, Nadkarni GN, Fleming F, Donovan MJ, Fields R. Real World Evidence and Clinical Utility of KidneyIntelX on Patients With Early-Stage Diabetic Kidney Disease: Interim Results on Decision Impact and Outcomes. J Prim Care Community Health 2022; 13:21501319221138196. [PMID: 36404761 PMCID: PMC9677284 DOI: 10.1177/21501319221138196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION AND OBJECTIVE The lack of precision to identify patients with early-stage diabetic kidney disease (DKD) at near-term risk for progressive decline in kidney function results in poor disease management often leading to kidney failure requiring unplanned dialysis. The KidneyIntelX is a multiplex, bioprognostic, immunoassay consisting of 3 plasma biomarkers and clinical variables that uses machine learning to generate a risk score for progressive decline in kidney function over 5-year in adults with early-stage DKD. Our objective was to assess the impact of KidneyIntelX on management and outcomes in a Health System in the real-world evidence (RWE) study. METHODS KidneyIntelX was introduced into a large metropolitan Health System via a population health-defined approved care pathway for patients with stages 1 to 3 DKD between [November 2020 to March 2022]. Decision impact on visit frequency, medication management, specialist referral, and selected lab values was assessed. We performed an interim analysis in patients through 6-months post-test date to evaluate the impact of risk level with clinical decision-making and outcomes. RESULTS A total of 1686 patients were enrolled in the RWE study and underwent KidneyIntelX testing and subsequent care pathway management. The median age was 68 years, 52% were female, 26% self-identified as Black, and 94% had hypertension. The median baseline eGFR was 59 ml/minute/1.73 m2, urine albumin-creatinine ratio was 69 mg/g, and HbA1c was 7.7%. After testing, a clinical encounter in the first month occurred in 13%, 43%, and 53% of low-risk, intermediate-risk, and high-risk patients, respectively and 46%, 61%, and 71% had at least 1 action taken within the first 6 months. High-risk patients were more likely to be placed on SGLT2 inhibitors (OR = 4.56; 95% CI 3.00-6.91 vs low-risk), and more likely to be referred to a specialist such as a nephrologist, endocrinologist, or dietician (OR = 2.49; 95% CI 1.53-4.01) compared to low-risk patients. CONCLUSIONS The combination of KidneyIntelX, clinical guidelines and educational support resulted in changes in clinical management by clinicians. After testing, there was an increase in visit frequency, referrals for disease management, and introduction to guideline-recommended medications. These differed by risk category, indicating an impact of KidneyIntelX risk stratification on clinical care.
Collapse
Affiliation(s)
- Joji Tokita
- The Barbara T Murphy Division of
Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aida Vega
- Department of General Internal Medicine
at Mount Sinai, New York, NY, USA
| | | | - Nidhi Naik
- Icahn School of Medicine at Mount
Sinai, New York, NY, USA
| | - Shivani Rathi
- Icahn School of Medicine at Mount
Sinai, New York, NY, USA
| | | | - Stephanie Wang
- Department of General Internal Medicine
at Mount Sinai, New York, NY, USA
| | - Leonard Amoruso
- Department of General Internal Medicine
at Mount Sinai, New York, NY, USA
| | | | - Steven G. Coca
- The Barbara T Murphy Division of
Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Girish N. Nadkarni
- The Barbara T Murphy Division of
Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Michael J. Donovan
- Icahn School of Medicine at Mount
Sinai, New York, NY, USA
- Renalytix AI, Inc, New York, NY,
USA
| | | |
Collapse
|
75
|
Wu TH, Chang LH, Chu CH, Hwu CM, Chen HS, Lin LY. Soluble tumor necrosis factor receptor 2 is associated with progressive diabetic kidney disease in patients with type 2 diabetes mellitus. PLoS One 2022; 17:e0266854. [PMID: 35413081 PMCID: PMC9004780 DOI: 10.1371/journal.pone.0266854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/21/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Chronic low-grade inflammation is considered one of the major mechanisms for the progression of diabetic kidney disease. We investigated the prognostic value of circulating soluble tumor necrosis factor receptor 2 (sTNFR2) for early nephropathy in patients with type 2 diabetes. MATERIALS AND METHODS A total of 364 patients with type 2 diabetes and an estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73m2 were followed up for a median of 4 years. Renal outcomes were defined as a composite of either or both a >30% decline in the eGFR and/or albuminuria stage progression determined with consecutive tests. RESULTS Seventy-three patients developed renal composite events. Serum concentrations of sTNFR2 were strongly associated with the risk of renal function decline and progressive changes in albuminuria. Through a receiver operating characteristic curve analysis, a serum sTNFR2 level of 1.608 ng/mL was adopted as the discriminator value for predicting renal outcomes (area under the curve 0.63, 95% confidence interval 0.57-0.70, p < 0.001), yielding a sensitivity of 75.3% and a specificity of 51.2%. The association of sTNFR2 levels ≥1.608 ng/mL to renal outcomes was significant after adjusting for relevant variables (hazard ratio 2.27, 95% confidence interval 1.23-4.20, p = 0.009) and remained consistent across subgroups stratified by age, sex, systolic blood pressure, eGFR, albuminuria, and the use of renin-angiotensin system blockers. CONCLUSIONS Higher circulating levels of sTNFR2 are independently associated with an eGFR decline and progressive albuminuria in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Tsung-Hui Wu
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Hsin Chang
- Division of Endocrinology and Metabolism, Department of Medicine, Yeezen General Hospital, Taoyuan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Chia-Huei Chu
- Department of Otorhinolaryngology-Head and Neck Surgery, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Audiology and Speech Language Pathology, Mackay Medical College, New Taipei City, Taiwan
| | - Chii-Min Hwu
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Harn-Shen Chen
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Liang-Yu Lin
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
76
|
Tu C, Wang L, Wei L. The Role of PKM2 in Diabetic Microangiopathy. Diabetes Metab Syndr Obes 2022; 15:1405-1412. [PMID: 35548702 PMCID: PMC9081029 DOI: 10.2147/dmso.s366403] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetic microangiopathy is among the most common complications affecting patients with diabetes, and includes both diabetic retinopathy (DR) and diabetic nephropathy (DKD). Diabetic microangiopathy remains a persistent threat to the health and quality of life of affected patients. Mechanistically, the severity of DR and DKD is tied to mitochondrial and glucose metabolism abnormalities, with the activation of the glycolytic enzyme pyruvate kinase M2 (PKM2) contributing to mitochondrial and glomerular dysfunction, abnormal renal hemodynamics, and retinopathy. PKM2 can activate inflammatory bodies in macrophages to promote the release of inflammatory mediators, and serves as a key regulator of inflammatory factors, chemokines and adhesion molecules. As such, there is sufficient evidence that PKM2 can be used as a biomarker for the diagnosis of diabetes and diabetic microangiopathy. Here, we survey the mechanisms whereby PKM2 contributes to diabetes-related microvascular diseases, associated regulatory roles, post-translational modifications, and the potential utility of PKM2 as a therapeutic target. Through this literature review, we have determined that PKM2 offers promise as both a diagnostic marker and therapeutic target with direct relevance to research pertaining to diabetic microangiopathy.
Collapse
Affiliation(s)
- Chao Tu
- Department of Internal Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Liangzhi Wang
- Department of Internal Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Lan Wei
- Department of Internal Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213000, People’s Republic of China
- Correspondence: Lan Wei, Department of Internal Medicine, the Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, Jiangsu, 213000, People’s Republic of China, Tel +86 0519 68871132, Email
| |
Collapse
|
77
|
Sur S, Nguyen M, Boada P, Sigdel TK, Sollinger H, Sarwal MM. FcER1: A Novel Molecule Implicated in the Progression of Human Diabetic Kidney Disease. Front Immunol 2021; 12:769972. [PMID: 34925339 PMCID: PMC8672419 DOI: 10.3389/fimmu.2021.769972] [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: 09/02/2021] [Accepted: 11/08/2021] [Indexed: 01/13/2023] Open
Abstract
Diabetic kidney disease (DKD) is a key microvascular complication of diabetes, with few therapies for targeting renal disease pathogenesis and progression. We performed transcriptional and protein studies on 103 unique blood and kidney tissue samples from patients with and without diabetes to understand the pathophysiology of DKD injury and its progression. The study was based on the use of 3 unique patient cohorts: peripheral blood mononuclear cell (PBMC) transcriptional studies were conducted on 30 patients with DKD with advancing kidney injury; Gene Expression Omnibus (GEO) data was downloaded, containing transcriptional measures from 51 microdissected glomerulous from patients with DKD. Additionally, 12 independent kidney tissue sections from patients with or without DKD were used for validation of target genes in diabetic kidney injury by kidney tissue immunohistochemistry and immunofluorescence. PBMC DKD transcriptional analysis, identified 853 genes (p < 0.05) with increasing expression with progression of albuminuria and kidney injury in patients with diabetes. GEO data was downloaded, normalized, and analyzed for significantly changed genes. Of the 325 significantly up regulated genes in DKD glomerulous (p < 0.05), 28 overlapped in PBMC and diabetic kidney, with perturbed FcER1 signaling as a significantly enriched canonical pathway. FcER1 was validated to be significantly increased in advanced DKD, where it was also seen to be specifically co-expressed in the kidney biopsy with tissue mast cells. In conclusion, we demonstrate how leveraging public and private human transcriptional datasets can discover and validate innate immunity and inflammation as key mechanistic pathways in DKD progression, and uncover FcER1 as a putative new DKD target for rational drug design.
Collapse
Affiliation(s)
- Swastika Sur
- Division of Transplant Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Mark Nguyen
- Division of Transplant Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Patrick Boada
- Division of Transplant Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Tara K Sigdel
- Division of Transplant Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Hans Sollinger
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Minnie M Sarwal
- Division of Transplant Surgery, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
78
|
Casagrande V, Federici M, Menghini R. TIMP3 involvement and potentiality in the diagnosis, prognosis and treatment of diabetic nephropathy. Acta Diabetol 2021; 58:1587-1594. [PMID: 34181080 PMCID: PMC8542557 DOI: 10.1007/s00592-021-01766-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/14/2021] [Indexed: 11/28/2022]
Abstract
Diabetic kidney disease, one of the most severe complications associated with diabetes, is characterized by albuminuria, glomerulosclerosis and progressive loss of renal function. Loss of TIMP3, an Extracellular matrix-bound protein, is a hallmark of diabetic nephropathy in human and mouse models, suggesting its pivotal role in renal diseases associated to diabetes. There is currently no specific therapy for diabetic nephropathy, and the ability to restore high TIMP3 activity specifically in the kidney may represent a potential therapeutic strategy for the amelioration of renal injury under conditions in which its reduction is directly related to the disease. Increasing evidence shows that diabetic nephropathy is also regulated by epigenetic mechanisms, including noncoding RNA. This review recapitulates the pathological, diagnostic and therapeutic potential roles of TIMP3 and the noncoding RNA (microRNA, long noncoding RNA) related to its expression, in the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Viviana Casagrande
- Departments of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Massimo Federici
- Departments of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Center for Atherosclerosis, Department of Medical Sciences, Policlinico Tor Vergata University, Rome, Italy
| | - Rossella Menghini
- Departments of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
79
|
Chang LH, Hwu CM, Chu CH, Lin YC, Huang CC, You JY, Chen HS, Lin LY. The combination of soluble tumor necrosis factor receptor type 1 and fibroblast growth factor 21 exhibits better prediction of renal outcomes in patients with type 2 diabetes mellitus. J Endocrinol Invest 2021; 44:2609-2619. [PMID: 33834419 DOI: 10.1007/s40618-021-01568-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE Numerous biomarkers of diabetic kidney disease (DKD) are associated with renal prognosis but head-to-head comparisons are lacking. This study aimed to examine the association of soluble tumor necrosis factor receptor type 1 (sTNFR1), fibroblast growth factor 21 (FGF-21), endocan, N-terminal pro-brain natriuretic peptide (NT-pro-BNP), and renal outcomes of patients with or without clinical signs of DKD. METHODS A total of 312 patients were enrolled in a prospective observational study that excluded individuals with estimated glomerular filtration rates (eGFR) < 30 mL/min/1.73 m2. Composite renal outcomes included either a > 30% decline in eGFR and worsening albuminuria or both from consecutive tests of blood/urine during a 3.5-year follow-up period. RESULTS Higher sTNFR1 and FGF-21, rather than endocan and NT-pro-BNP, levels were associated with renal outcomes but the significance was lost after adjusting for confounders. However, sTNFR1 levels ≥ 9.79 pg/dL or FGF-21 levels ≥ 1.40 pg/dL were associated with renal outcomes after adjusting for the confounders (hazard ration [HR] 2.76, 95% confidence interval [CI] 1.36-5.60, p = 0.005 for sTNFR1 level; HR 1.95, 95% CI 1.03-3.69, p = 0.03 for FGF-21 level). The combination of both levels exhibited even better association with renal outcomes than did either one alone (adjusted HR 4.45, 95% CI 1.86-10.65, p = 0.001). The results were consistent among patients with preserved renal function and normoalbuminuria. CONCLUSION Both sTNFR1 and FGF-21 levels were associated with renal outcomes of in patients with type 2 diabetes, and the combination of the abovementioned markers exhibits better predictability.
Collapse
Affiliation(s)
- L-H Chang
- Division of Endocrinology and Metabolism, Department of Medicine, Yeezen General Hospital, Taoyuan, Taiwan
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - C-M Hwu
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - C-H Chu
- Department of Otorhinolaryngology-Head and Neck Surgery, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Audiology and Speech Language Pathology, Mackay Medical College, New Taipei City, Taiwan
| | - Y-C Lin
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - C-C Huang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - J-Y You
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - H-S Chen
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - L-Y Lin
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
80
|
Yang Y, Shi K, Patel DM, Liu F, Wu T, Chai Z. How to inhibit transforming growth factor beta safely in diabetic kidney disease. Curr Opin Nephrol Hypertens 2021; 30:115-122. [PMID: 33229911 DOI: 10.1097/mnh.0000000000000663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW Diabetic kidney disease (DKD) is a leading cause of mortality and morbidity in diabetes. This review aims to discuss the major features of DKD, to identify the difficult barrier encountered in developing a therapeutic strategy and to provide a potentially superior novel approach to retard DKD. RECENT FINDINGS Renal inflammation and fibrosis are prominent features of DKD. Transforming growth factor beta (TGFβ) with its activity enhanced in DKD plays a key pathological profibrotic role in promoting renal fibrosis. However, TGFβ is a difficult drug target because it has multiple important physiological functions, such as immunomodulation. These physiological functions of TGFβ can be interrupted as a result of complete blockade of the TGFβ pathway if TGFβ is directly targeted, leading to catastrophic side-effects, such as fulminant inflammation. Cell division autoantigen 1 (CDA1) is recently identified as an enhancer of profibrotic TGFβ signaling and inhibitor of anti-inflammatory SIRT1. Renal CDA1 expression is elevated in human DKD as well as in rodent models of DKD. Targeting CDA1, by either genetic approach or pharmacological approach in mice, leads to concurrent attenuation of renal fibrosis and inflammation without any deleterious effects observed. SUMMARY Targeting CDA1, instead of directly targeting TGFβ, represents a superior approach to retard DKD.
Collapse
Affiliation(s)
- Yuxin Yang
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Pathology, Zunyi maternity and Child Healthcare Hospital, Zunyi
| | - Kexin Shi
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Devang M Patel
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Fang Liu
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Tieqiao Wu
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Zhonglin Chai
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
81
|
Connolly P, Stapleton S, Mosoyan G, Fligelman I, Tonar YC, Fleming F, Donovan MJ. Analytical validation of a multi-biomarker algorithmic test for prediction of progressive kidney function decline in patients with early-stage kidney disease. Clin Proteomics 2021; 18:26. [PMID: 34789168 PMCID: PMC8597271 DOI: 10.1186/s12014-021-09332-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/28/2021] [Indexed: 04/03/2023] Open
Abstract
Background The KidneyIntelX™ test applies a machine learning algorithm that incorporates plasma biomarkers and clinical variables to produce a composite risk score to predict a progressive decline in kidney function in patients with type 2 diabetes (T2D) and early-stage chronic kidney disease (CKD). The following studies describe the analytical validation of the KidneyIntelX assay including impact of observed methodologic variability on the composite risk score. Methods Analytical performance studies of sensitivity, precision, and linearity were performed on three biomarkers assayed in multiplexed format: kidney injury molecule-1 (KIM-1), soluble tumor necrosis factor receptor-1 (sTNFR-1) and soluble tumor necrosis factor receptor-2 (sTNFR-2) based on Clinical Laboratory Standards Institute (CLSI) guidelines. Analytical variability across twenty (20) experiments across multiple days, operators, and reagent lots was assessed to examine the impact on the reproducibility of the composite risk score. Analysis of cross-reactivity and interfering substances was also performed. Results Assays for KIM-1, sTNFR-1 and sTNFR-2 demonstrated acceptable sensitivity. Mean within-laboratory imprecision coefficient of variation (CV) was established as less than 9% across all assays in a multi-lot study. The linear range of the assays was determined as 12–5807 pg/mL, 969–23,806 pg/mL and 4256–68,087 pg/mL for KIM-1, sTNFR-1 and sTNFR-2, respectively. The average risk score CV% was less than 5%, with 98% concordance observed for assignment of risk categories. Cross-reactivity between critical assay components in a multiplexed format did not exceed 1.1%. Conclusions The set of analytical validation studies demonstrated robust analytical performance across all three biomarkers contributing to the KidneyIntelX risk score, meeting or exceeding specifications established during characterization studies. Notably, reproducibility of the composite risk score demonstrated that expected analytical laboratory variation did not impact the assigned risk category, and therefore, the clinical validity of the reported results. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09332-y.
Collapse
|
82
|
Ceccarelli Ceccarelli D, Paleari R, Solerte B, Mosca A. Re-thinking diabetic nephropathy: Microalbuminuria is just a piece of the diagnostic puzzle. Clin Chim Acta 2021; 524:146-153. [PMID: 34767792 DOI: 10.1016/j.cca.2021.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/29/2021] [Accepted: 11/07/2021] [Indexed: 12/13/2022]
Abstract
The decline of the estimated glomerular filtration rate (eGFR) and the presence of albuminuria are the typical hallmarks of kidney disease arising as one of the most frequent diabetic complications over a long period of time, generally known as diabetic nephropathy or diabetes kidney disease (DKD). However, a decline in the renal function may occur in diabetic patients for other reasons unrelated to glycemic control, and this condition is known as non-diabetic kidney disease (NDKD). In this opinion paper we will review these conditions, and we outline the importance of other investigations, such as kidney biopsy and the measurement of novel biomarkers, in order to identify the disease progression early, and to allow a timely intervention. We will also focus on the actual limits of the quantitative measurements of albumin in urine, especially with regards to potential interferences due to the treatment of patients with statins.
Collapse
Affiliation(s)
| | - Renata Paleari
- Dip. di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milano, Italy
| | - Bruno Solerte
- Dip. di Medicina Interna e Terapia Medica, Università degli Studi di Pavia, Pavia, Italy
| | - Andrea Mosca
- Dip. di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
83
|
Mohamed RH, Sedky AA, Hamam GG, Elkhateb L, Kamar SA, Adel S, Tawfik SS. Sitagliptin's renoprotective effect in a diabetic nephropathy model in rats: The potential role of PI3K/AKT pathway. Fundam Clin Pharmacol 2021; 36:324-337. [PMID: 34735026 DOI: 10.1111/fcp.12736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022]
Abstract
Management of diabetic nephropathy (DN) is far from satisfactory. There is a rising role of the involvement of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway in the pathogenesis of DN. This study aimed at investigating the renoprotective effects of PI3K/AKT pathway via sitagliptin in a rat model of DN. Thirty-two male Wistar rats were divided into four groups (eight rats each): (I) control, (II) sitagliptin, (III) DN, and (IV) DN + sitagliptin. Fasting blood glucose (FBG), kidney index, and kidney function tests in both blood and urine were measured. The levels of superoxide dismutase (SOD), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta (TGF-β) and gene expressions of PI3K, pPI3K, AKT, and pAKT in renal tissue were detected. Renal histopathological and immunohistochemical studies were evaluated. DN + sitagliptin group showed significant decrease in FBG and kidney index, improvement in kidney function tests, and a decrease in levels of TNF-α and TGF-β in renal tissues compared with DN group. This was associated with significant increase in SOD and gene expressions of PI3K and AKT and their phosphorylated active forms in renal tissue in DN + sitagliptin group compared with DN group. Moreover, DN + sitagliptin group showed apparent decrease in amount of collagen fibers and expression of alpha-smooth muscle actin (α-SMA) compared with DN group. This work shows that sitagliptin improved renal functions and histopathological changes, impeded inflammation, and oxidative stress and upregulated PI3K/AKT pathway which highlights its renoprotective effects in a rat model of DN.
Collapse
Affiliation(s)
- Reham Hussein Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amina Ahmed Sedky
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ghada Galal Hamam
- Department of Histology and cell biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Lobna Elkhateb
- Department of Histology and cell biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherif A Kamar
- Department of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Seham Adel
- Department of Biochemistry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherin Shafik Tawfik
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
84
|
Trajectories of kidney function in diabetes: a clinicopathological update. Nat Rev Nephrol 2021; 17:740-750. [PMID: 34363037 DOI: 10.1038/s41581-021-00462-y] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Diabetic nephropathy has been traditionally diagnosed based on persistently high albuminuria and a subsequent decline in glomerular filtration rate (GFR), which is widely recognized as the classical phenotype of diabetic kidney disease (DKD). Several studies have emphasized that trajectories of kidney function in patients with diabetes (specifically, changes in GFR and albuminuria over time) can differ from this classical DKD phenotype. Three alternative DKD phenotypes have been reported to date and are characterized by albuminuria regression, a rapid decline in GFR, or non-proteinuric or non-albuminuric DKD. Although kidney biopsies are not typically required for the diagnosis of DKD, a few studies of biopsy samples from patients with DKD have demonstrated that changes in kidney function associate with specific histopathological findings in diabetes. In addition, various clinical and biochemical parameters are related to trajectories of GFR and albuminuria. Collectively, pathological and clinical characteristics can be used to predict trajectories of GFR and albuminuria in diabetes. Furthermore, cohort studies have suggested that the risks of kidney and cardiovascular outcomes might vary among different phenotypes of DKD. A broader understanding of the clinical course of DKD is therefore crucial to improve risk stratification and enable early interventions that prevent adverse outcomes.
Collapse
|
85
|
Ishii H, Kaneko S, Yanai K, Aomatsu A, Hirai K, Ookawara S, Morishita Y. MicroRNA Expression Profiling in Diabetic Kidney Disease. Transl Res 2021; 237:31-52. [PMID: 34102327 DOI: 10.1016/j.trsl.2021.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 12/29/2022]
Abstract
The microRNAs (miRNAs) that can regulate diabetic kidney disease (DKD) have not been fully characterized. The aim of this study was to identify the miRNAs that affect DKD and could be used as specific biomarkers or therapeutic agents. First, kidney tissues from two DKD mouse models and control mice were screened for differences in miRNA expression by microarray analysis followed by quantitative real-time reverse transcription-PCR. Six miRNAs were differentially expressed from controls in both DKD mouse models. Among them, miRNA-125b-5p and miRNA-181b-5p were exclusively downregulated in the DKD mouse model. Next, we administered miRNA-181b-5p-mimic to DKD mice, which reduced the albuminuria and abnormal mesangial expansion. Pathway analysis and database research revealed that overexpression of miRNA-181b-5p significantly altered the expression of seven mRNAs in six known signaling pathways in the kidneys of DKD mice. Furthermore, the serum level of miRNA-125b-5p was significantly higher in patients with DKD (1.89±0.40-fold, P<0.05) compared with patients with other kidney diseases (0.94±0.13-fold) and healthy subjects (1.00±0.19-fold). Serum levels of miRNA-181b-5p were lower in patients with DKD (0.30±0.06-fold, P<0.05) compared with patients with other kidney diseases (1.06±0.20-fold) and healthy subjects (1.00±0.16-fold). These results suggest that miRNA-125b-5p and miRNA-181b-5p may represent novel diagnostic biomarkers and that miRNA-181b-5p may represent a therapeutic target for DKD.
Collapse
Affiliation(s)
- Hiroki Ishii
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Shohei Kaneko
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Katsunori Yanai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Akinori Aomatsu
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Keiji Hirai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Susumu Ookawara
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yoshiyuki Morishita
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan.
| |
Collapse
|
86
|
Saputro SA, Pattanaprateep O, Pattanateepapon A, Karmacharya S, Thakkinstian A. Prognostic models of diabetic microvascular complications: a systematic review and meta-analysis. Syst Rev 2021; 10:288. [PMID: 34724973 PMCID: PMC8561867 DOI: 10.1186/s13643-021-01841-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Many prognostic models of diabetic microvascular complications have been developed, but their performances still varies. Therefore, we conducted a systematic review and meta-analysis to summarise the performances of the existing models. METHODS Prognostic models of diabetic microvascular complications were retrieved from PubMed and Scopus up to 31 December 2020. Studies were selected, if they developed or internally/externally validated models of any microvascular complication in type 2 diabetes (T2D). RESULTS In total, 71 studies were eligible, of which 32, 30 and 18 studies initially developed prognostic model for diabetic retinopathy (DR), chronic kidney disease (CKD) and end stage renal disease (ESRD) with the number of derived equations of 84, 96 and 51, respectively. Most models were derived-phases, some were internal and external validations. Common predictors were age, sex, HbA1c, diabetic duration, SBP and BMI. Traditional statistical models (i.e. Cox and logit regression) were mostly applied, otherwise machine learning. In cohorts, the discriminative performance in derived-logit was pooled with C statistics of 0.82 (0.73‑0.92) for DR and 0.78 (0.74‑0.83) for CKD. Pooled Cox regression yielded 0.75 (0.74‑0.77), 0.78 (0.74‑0.82) and 0.87 (0.84‑0.89) for DR, CKD and ESRD, respectively. External validation performances were sufficiently pooled with 0.81 (0.78‑0.83), 0.75 (0.67‑0.84) and 0.87 (0.85‑0.88) for DR, CKD and ESRD, respectively. CONCLUSIONS Several prognostic models were developed, but less were externally validated. A few studies derived the models by using appropriate methods and were satisfactory reported. More external validations and impact analyses are required before applying these models in clinical practice. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42018105287.
Collapse
Affiliation(s)
- Sigit Ari Saputro
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Pyathai, Bangkok, 10400, Thailand.,Department of Epidemiology Biostatistics Population and Health Promotion, Faculty of Public Health, Airlangga University, Surabaya, Indonesia
| | - Oraluck Pattanaprateep
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Pyathai, Bangkok, 10400, Thailand.
| | - Anuchate Pattanateepapon
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Pyathai, Bangkok, 10400, Thailand
| | - Swekshya Karmacharya
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Pyathai, Bangkok, 10400, Thailand
| | - Ammarin Thakkinstian
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Pyathai, Bangkok, 10400, Thailand
| |
Collapse
|
87
|
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) is a silent disease, causing significant health and economic burden worldwide. It is of strong clinical value to identify novel prognostic, predictive, and pharmacodynamic biomarkers of kidney function, as current available measures have limitations. We reviewed the advances in biomarkers in CKD over the preceding year. RECENT FINDINGS The most frequently studied prognostic plasma biomarkers during recent year were plasma TNFR1, TNFR2, KIM1 and urinary MCP-1 and EGF. New biomarkers such as plasma WFDC2, MMP-7, EFNA4, EPHA2 may also have potential to serve as prognostic biomarkers. There is a shortage of data on biomarkers that are predictive of response to treatments. Data on novel biomarkers to serve as pharmacodynamic biomarkers are limited, but there are emerging data that plasmaTNFR1, TNFR2, KIM-1 are not only prognostic at baseline, but can also contribute to time-updated response signals in response to therapy. SUMMARY Data continue to emerge on applicable biomarkers for prognostic clinical risk stratification, prediction of therapeutic response and assessment of early efficacy of interventions. Although more studies are needed for refinement and specific clinical utility, there seems to be sufficient data to support clinical implementation for some biomarkers.
Collapse
Affiliation(s)
- Azadeh Zabetian
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | |
Collapse
|
88
|
Sen T, Li J, Neuen BL, Neal B, Arnott C, Parikh CR, Coca SG, Perkovic V, Mahaffey KW, Yavin Y, Rosenthal N, Hansen MK, Heerspink HJL. Effects of the SGLT2 inhibitor canagliflozin on plasma biomarkers TNFR-1, TNFR-2 and KIM-1 in the CANVAS trial. Diabetologia 2021; 64:2147-2158. [PMID: 34415356 PMCID: PMC8423682 DOI: 10.1007/s00125-021-05512-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/19/2021] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Higher plasma concentrations of tumour necrosis factor receptor (TNFR)-1, TNFR-2 and kidney injury molecule-1 (KIM-1) have been found to be associated with higher risk of kidney failure in individuals with type 2 diabetes in previous studies. Whether drugs can reduce these biomarkers is not well established. We measured these biomarkers in samples of the CANVAS study and examined the effect of the sodium-glucose cotransporter 2 inhibitor canagliflozin on these biomarkers and assessed whether the early change in these biomarkers predict cardiovascular and kidney outcomes in individuals with type 2 diabetes in the CANagliflozin cardioVascular Assessment Study (CANVAS). METHODS Biomarkers were measured with immunoassays (proprietary multiplex assay performed by RenalytixAI, New York, NY, USA) at baseline and years 1, 3 and 6. Mixed-effects models for repeated measures assessed the effect of canagliflozin vs placebo on the biomarkers. Associations of baseline levels and the early change (baseline to year 1) for each biomarker with the kidney outcome were assessed using multivariable-adjusted Cox regression. RESULTS In total, 3523/4330 (81.4%) of the CANVAS participants had available samples at baseline. Each doubling in baseline TNFR-1, TNFR-2 and KIM-1 was associated with a higher risk of kidney outcomes, with corresponding HRs of 3.7 (95% CI 2.3, 6.1; p < 0.01), 2.7 (95% CI 2.0, 3.6; p < 0.01) and 1.5 (95% CI 1.2, 1.8; p < 0.01), respectively. Canagliflozin reduced the level of the plasma biomarkers with differences in TNFR-1, TNFR-2 and KIM-1 between canagliflozin and placebo during follow-up of 2.8% (95% CI 3.4%, 1.3%; p < 0.01), 1.9% (95% CI 3.5%, 0.2%; p = 0.03) and 26.7% (95% CI 30.7%, 22.7%; p < 0.01), respectively. Within the canagliflozin treatment group, each 10% reduction in TNFR-1 and TNFR-2 at year 1 was associated with a lower risk of the kidney outcome (HR 0.8 [95% CI 0.7, 1.0; p = 0.02] and 0.9 [95% CI 0.9, 1.0; p < 0.01] respectively), independent of other patient characteristics. The baseline and 1 year change in biomarkers did not associate with cardiovascular or heart failure outcomes. CONCLUSIONS/INTERPRETATION Canagliflozin decreased KIM-1 and modestly reduced TNFR-1 and TNFR-2 compared with placebo in individuals with type 2 diabetes in CANVAS. Early decreases in TNFR-1 and TNFR-2 during canagliflozin treatment were independently associated with a lower risk of kidney disease progression, suggesting that TNFR-1 and TNFR-2 have the potential to be pharmacodynamic markers of response to canagliflozin.
Collapse
Affiliation(s)
- Taha Sen
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Jingwei Li
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW, Australia
| | - Brendon L Neuen
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW, Australia
| | - Bruce Neal
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW, Australia
| | - Clare Arnott
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW, Australia
| | | | - Steven G Coca
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vlado Perkovic
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW, Australia
| | - Kenneth W Mahaffey
- Stanford Center for Clinical Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Yshai Yavin
- Janssen Research & Development LLC, Spring House, PA, USA
| | | | | | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, the Netherlands.
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
89
|
Greenberg JH, Abraham AG, Xu Y, Schelling JR, Feldman HI, Sabbisetti VS, Ix JH, Jogalekar MP, Coca S, Waikar SS, Shlipak MG, Warady BA, Vasan RS, Kimmel PL, Bonventre JV, Denburg M, Parikh CR, Furth S. Urine Biomarkers of Kidney Tubule Health, Injury, and Inflammation are Associated with Progression of CKD in Children. J Am Soc Nephrol 2021; 32:2664-2677. [PMID: 34544821 PMCID: PMC8722795 DOI: 10.1681/asn.2021010094] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/28/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Novel urine biomarkers may improve identification of children at greater risk of rapid kidney function decline, and elucidate the pathophysiology of CKD progression. METHODS We investigated the relationship between urine biomarkers of kidney tubular health (EGF and α-1 microglobulin), tubular injury (kidney injury molecule-1; KIM-1), and inflammation (monocyte chemoattractant protein-1 [MCP-1] and YKL-40) and CKD progression. The prospective CKD in Children Study enrolled children aged 6 months to 16 years with an eGFR of 30-90ml/min per 1.73m2. Urine biomarkers were assayed a median of 5 months [IQR: 4-7] after study enrollment. We indexed the biomarker to urine creatinine by dividing the urine biomarker concentration by the urine creatinine concentration to account for the concentration of the urine. The primary outcome was CKD progression (a composite of a 50% decline in eGFR or kidney failure) during the follow-up period. RESULTS Overall, 252 of 665 children (38%) reached the composite outcome over a median follow-up of 6.5 years. After adjustment for covariates, children with urine EGF concentrations in the lowest quartile were at a seven-fold higher risk of CKD progression versus those with concentrations in the highest quartile (fully adjusted hazard ratio [aHR], 7.1; 95% confidence interval [95% CI], 3.9 to 20.0). Children with urine KIM-1, MCP-1, and α-1 microglobulin concentrations in the highest quartile were also at significantly higher risk of CKD progression versus those with biomarker concentrations in the lowest quartile. Addition of the five biomarkers to a clinical model increased the discrimination and reclassification for CKD progression. CONCLUSIONS After multivariable adjustment, a lower urine EGF concentration and higher urine KIM-1, MCP-1, and α-1 microglobulin concentrations were each associated with CKD progression in children.
Collapse
Affiliation(s)
- Jason H. Greenberg
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut,Department of Medicine Clinical and Translational Research Accelerator, Yale University School of Medicine, New Haven, Connecticut
| | - Alison G. Abraham
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Yunwen Xu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jeffrey R. Schelling
- Department of Internal Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Harold I. Feldman
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Joachim H. Ix
- Division of Nephrology-Hypertension, University of California San Diego, San Diego, California,Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, California
| | - Manasi P. Jogalekar
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Steven Coca
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sushrut S. Waikar
- Section of Nephrology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Michael G. Shlipak
- UCSF Division of General Internal Medicine at the VA, Kidney Health Research Collaborative, San Francisco Veterans Affairs Health Care System and University of California, San Francisco, California
| | - Bradley A. Warady
- Department of Pediatrics, Children’s Mercy Kansas City, Kansas City, Missouri
| | - Ramachandran S. Vasan
- Departments of Medicine and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Paul L. Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Joseph V. Bonventre
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Michelle Denburg
- Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Chirag R. Parikh
- Department of Internal Medicine, Johns Hopkins School of Medicine, Baltimore, New York
| | - Susan Furth
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | | | | |
Collapse
|
90
|
Feng ST, Yang Y, Yang JF, Gao YM, Cao JY, Li ZL, Tang TT, Lv LL, Wang B, Wen Y, Sun L, Xing GL, Liu BC. Urinary sediment CCL5 messenger RNA as a potential prognostic biomarker of diabetic nephropathy. Clin Kidney J 2021; 15:534-544. [PMID: 35211307 PMCID: PMC8862108 DOI: 10.1093/ckj/sfab186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
ABSTRACT
Background
Urinary sediment messenger RNAs (mRNAs) have been shown as novel biomarkers of kidney disease. We aimed to identify targeted urinary mRNAs in diabetic nephropathy (DN) based on bioinformatics analysis and clinical validation.
Methods
Microarray studies of DN were searched in the GEO database and Nephroseq platform. Gene modules negatively correlated with estimated glomerular filtration rate (eGFR) were identified by informatics methods. Hub genes were screened within the selected modules. In validation cohorts, a quantitative polymerase chain reaction assay was used to compare the expression levels of candidate mRNAs. Patients with renal biopsy–confirmed DN were then followed up for a median time of 21 months. End-stage renal disease (ESRD) was defined as the primary endpoint. Multivariate Cox proportional hazards regression was developed to evaluate the prognostic values of candidate mRNAs.
Results
Bioinformatics analysis revealed four chemokines (CCL5, CXCL1, CXLC6 and CXCL12) as candidate mRNAs negatively correlated with eGFR, of which CCL5 and CXCL1 mRNA levels were upregulated in the urinary sediment of patients with DN. In addition, urinary sediment mRNA of CXCL1 was negatively correlated with eGFR (r = −0.2275, P = 0.0301) and CCL5 level was negatively correlated with eGFR (r = −0.4388, P < 0.0001) and positively correlated with urinary albumin:creatinine ratio (r = 0.2693, P = 0.0098); also, CCL5 and CXCL1 were upregulated in patients with severe renal interstitial fibrosis. Urinary sediment CCL5 mRNA was an independent predictor of ESRD [hazard ratio 1.350 (95% confidence interval 1.045–1.745)].
Conclusions
Urinary sediment CCL5 and CXCL1 mRNAs were upregulated in DN patients and associated with a decline in renal function and degree of renal interstitial fibrosis. Urinary sediment CCL5 mRNA could be used as a potential prognostic biomarker of DN.
Collapse
Affiliation(s)
- Song-Tao Feng
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Yang Yang
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jin-Fei Yang
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Yue-Ming Gao
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Jing-Yuan Cao
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Yi Wen
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Lin Sun
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Guo-Lan Xing
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
91
|
Tumor Necrosis Receptor Superfamily Interact with Fusion and Fission of Mitochondria of Adipose Tissue in Obese Patients without Type 2 Diabetes. Biomedicines 2021; 9:biomedicines9091260. [PMID: 34572446 PMCID: PMC8470627 DOI: 10.3390/biomedicines9091260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
Interactions between receptors and ligands of the tumor necrosis factor superfamily (TNFSF) provide costimulatory signals that control the survival, proliferation, differentiation, and effector function of immune cells. All components of the TNF superfamily are associated with NF-kB functions that are not limited to cell death and may promote survival in the face of adipose tissue inflammation in obesity. Inflammation dysfunction of mitochondria is a key factor associated with insulin resistance in obesity. The aim of the study was to analyze the relationship of soluble forms of receptors and ligands of the TNF superfamily in blood plasma with mitochondrial dynamics in adipose tissue (greater omentum (GO) and subcutaneous adipose tissue (Sat)) of obese patients with and without type 2 diabetes mellitus (T2DM). Increased plasma sTNF-R1, sTNF-R2, sTNFRSF8 receptors, and ligands TNFSF12, TNFSF13, TNFSF13B are characteristic of obese patients without T2DM. The TNF-a levels in blood plasma were associated with a decrease in MFN2 gene expression in GO and IL-10 in blood plasma. The TNFSF12 levels contributed to a decrease in glucose levels, a decrease in BMI, and an increase in IL-10 levels by influencing the MFN2 gene expression in GO, which supports mitochondrial fusion.
Collapse
|
92
|
A Targeted Serum Metabolomics GC-MS Approach Identifies Predictive Blood Biomarkers for Retained Placenta in Holstein Dairy Cows. Metabolites 2021; 11:metabo11090633. [PMID: 34564449 PMCID: PMC8466882 DOI: 10.3390/metabo11090633] [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: 07/05/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/09/2023] Open
Abstract
The retained placenta is a common pathology of dairy cows. It is associated with a significant drop in the dry matter intake, milk yield, and increased susceptibility of dairy cows to metritis, mastitis, and displaced abomasum. The objective of this study was to identify metabolic alterations that precede and are associated with the disease occurrence. Blood samples were collected from 100 dairy cows at −8 and −4 weeks prior to parturition and on the day of retained placenta, and only 16 healthy cows and 6 cows affected by retained placenta were selected to measure serum polar metabolites by a targeted gas chromatography–mass spectroscopy (GC-MS) metabolomics approach. A total of 27 metabolites were identified and quantified in the serum. There were 10, 18, and 17 metabolites identified as being significantly altered during the three time periods studied. However, only nine metabolites were identified as being shared among the three time periods including five amino acids (Asp, Glu, Ser, Thr, and Tyr), one sugar (myo-inositol), phosphoric acid, and urea. The identified metabolites can be used as predictive biomarkers for the risk of retained placenta in dairy cows and might help explain the metabolic processes that occur prior to the incidence of the disease and throw light on the pathomechanisms of the disease.
Collapse
|
93
|
Vasquez-Rios G, Coca SG. Predictors of Kidney Disease Progression in Diabetes and Precision Medicine: Something Old, Something New, and Something Borrowed. J Am Soc Nephrol 2021; 32:2108-2111. [PMID: 34465605 PMCID: PMC8729853 DOI: 10.1681/asn.2021070945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- George Vasquez-Rios
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | |
Collapse
|
94
|
Yaqub S, Hamid A, Kashif W, Razzaque MRA, Farooque A, Ahmed B, Ram N. Rapidly progressive diabetic kidney disease: South Asian experience. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-021-00975-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
95
|
Gohda T, Yanagisawa N, Murakoshi M, Ueda S, Nishizaki Y, Nojiri S, Ohashi Y, Ohno I, Shibagaki Y, Imai N, Iimuro S, Kuwabara M, Hayakawa H, Kimura K, Hosoya T, Suzuki Y. Association Between Kidney Function Decline and Baseline TNFR Levels or Change Ratio in TNFR by Febuxostat Chiefly in Non-diabetic CKD Patients With Asymptomatic Hyperuricemia. Front Med (Lausanne) 2021; 8:634932. [PMID: 34322499 PMCID: PMC8310915 DOI: 10.3389/fmed.2021.634932] [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: 11/29/2020] [Accepted: 06/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The levels of circulating tumor necrosis factor receptor (TNFR) 1 and 2 help predict the future decline of estimated glomerular filtration rate (eGFR) chiefly in patients with diabetes. It has been recently reported that the change ratio in TNFR1 by SGLT2 inhibitor treatment is also related with future GFR decline in patients with diabetes. The aims of this study are to investigate the association between baseline TNFR levels and early change in TNFR levels by the non-purine selective xanthine oxidase inhibitor, febuxostat, and future eGFR decline chiefly in chronic kidney disease (CKD) patients without diabetes. Methods: We conducted a post-hoc analysis of the FEATHER study on patients with asymptomatic hyperuricemia and CKD stage 3, who were randomly assigned febuxostat 40 mg/day or matched placebo. This analysis included 426 patients in whom baseline stored samples were available. Serum TNFR levels at baseline were measured using enzyme-linked immunosorbent assay. Those levels were also measured using 12-week stored samples from 197 randomly selected patients. Results: Compared with placebo, short-term febuxostat treatment significantly decreased the median percent change from baseline in serum uric acid (−45.05, 95% CI −48.90 to −41.24 mg/dL), TNFR1 (1.10, 95% CI−2.25 to 4.40), and TNFR2 (1.66, 95% CI −1.72 to 4.93), but not TNFR levels. Over a median follow-up of 105 weeks, 30 patients (7.0%) experienced 30% eGFR decline from baseline. In the Cox multivariate model, high levels of baseline TNFR predicted a 30% eGFR decline, even after adjusting for age, sex, systolic blood pressure, high sensitivity C-reactive protein, uric acid, and presence or absence of febuxostat treatment and diabetes, in addition to baseline albumin to creatinine ratio and eGFR. Conclusion: Early change in circulating TNFR levels failed to predict future eGFR decline; however, regardless of febuxostat treatment, the elevated baseline level of TNFR was a strong predictor of 30% eGFR decline even in chiefly non-diabetic CKD patients with asymptomatic hyperuricemia.
Collapse
Affiliation(s)
- Tomohito Gohda
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | | - Maki Murakoshi
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Seiji Ueda
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuji Nishizaki
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Yasuo Ohashi
- Department of Integrated Science and Engineering for Sustainable Society, Chuo University, Tokyo, Japan
| | - Iwao Ohno
- Division of General Medicine, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naohiko Imai
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Satoshi Iimuro
- Innovation and Research Support Center, International University of Health and Welfare, Tokyo, Japan
| | - Masanari Kuwabara
- Intensive Care Unit, Department of Cardiology, Toranomon Hospital, Tokyo, Japan
| | | | | | - Tatsuo Hosoya
- Division of Chronic Kidney Disease Therapeutics, The Jikei University, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
96
|
Barutta F, Bellini S, Canepa S, Durazzo M, Gruden G. Novel biomarkers of diabetic kidney disease: current status and potential clinical application. Acta Diabetol 2021; 58:819-830. [PMID: 33528734 DOI: 10.1007/s00592-020-01656-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease (ESRD). Although both albuminuria and glomerular filtration rate (GFR) are well-established diagnostic/prognostic biomarkers of DKD, they have important limitations. There is, thus, increasing quest to find novel biomarkers to identify the disease in an early stage and to improve risk stratification. In this review, we will outline the major pitfalls of currently available markers, describe promising novel biomarkers, and discuss their potential clinical relevance. In particular, we will focus on the importance of recent advancements in multi-omic technologies in the discovery of new DKD biomarkers. In addition, we will provide an update on new emerging approaches to explore renal function and structure, using functional tests and imaging.
Collapse
Affiliation(s)
- Federica Barutta
- Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Stefania Bellini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Canepa
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Marilena Durazzo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Gabriella Gruden
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
97
|
Chan L, Nadkarni GN, Fleming F, McCullough JR, Connolly P, Mosoyan G, El Salem F, Kattan MW, Vassalotti JA, Murphy B, Donovan MJ, Coca SG, Damrauer SM. Derivation and validation of a machine learning risk score using biomarker and electronic patient data to predict progression of diabetic kidney disease. Diabetologia 2021; 64:1504-1515. [PMID: 33797560 PMCID: PMC8187208 DOI: 10.1007/s00125-021-05444-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022]
Abstract
AIM Predicting progression in diabetic kidney disease (DKD) is critical to improving outcomes. We sought to develop/validate a machine-learned, prognostic risk score (KidneyIntelX™) combining electronic health records (EHR) and biomarkers. METHODS This is an observational cohort study of patients with prevalent DKD/banked plasma from two EHR-linked biobanks. A random forest model was trained, and performance (AUC, positive and negative predictive values [PPV/NPV], and net reclassification index [NRI]) was compared with that of a clinical model and Kidney Disease: Improving Global Outcomes (KDIGO) categories for predicting a composite outcome of eGFR decline of ≥5 ml/min per year, ≥40% sustained decline, or kidney failure within 5 years. RESULTS In 1146 patients, the median age was 63 years, 51% were female, the baseline eGFR was 54 ml min-1 [1.73 m]-2, the urine albumin to creatinine ratio (uACR) was 6.9 mg/mmol, follow-up was 4.3 years and 21% had the composite endpoint. On cross-validation in derivation (n = 686), KidneyIntelX had an AUC of 0.77 (95% CI 0.74, 0.79). In validation (n = 460), the AUC was 0.77 (95% CI 0.76, 0.79). By comparison, the AUC for the clinical model was 0.62 (95% CI 0.61, 0.63) in derivation and 0.61 (95% CI 0.60, 0.63) in validation. Using derivation cut-offs, KidneyIntelX stratified 46%, 37% and 17% of the validation cohort into low-, intermediate- and high-risk groups for the composite kidney endpoint, respectively. The PPV for progressive decline in kidney function in the high-risk group was 61% for KidneyIntelX vs 40% for the highest risk strata by KDIGO categorisation (p < 0.001). Only 10% of those scored as low risk by KidneyIntelX experienced progression (i.e., NPV of 90%). The NRIevent for the high-risk group was 41% (p < 0.05). CONCLUSIONS KidneyIntelX improved prediction of kidney outcomes over KDIGO and clinical models in individuals with early stages of DKD.
Collapse
Affiliation(s)
- Lili Chan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Girish N Nadkarni
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fergus Fleming
- Renalytix AI Plc, Cardiff, UK
- Renalytix AI, Inc., New York, NY, USA
| | | | | | - Gohar Mosoyan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fadi El Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael W Kattan
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland, OH, USA
| | - Joseph A Vassalotti
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Barbara Murphy
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael J Donovan
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven G Coca
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
98
|
Natesan V, Kim SJ. Diabetic Nephropathy - a Review of Risk Factors, Progression, Mechanism, and Dietary Management. Biomol Ther (Seoul) 2021; 29:365-372. [PMID: 33888647 PMCID: PMC8255138 DOI: 10.4062/biomolther.2020.204] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/18/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) leads to many health problems like diabetic nephropathy (DN). One of the key factors for chronic kidney disease and end-stage renal disease (ESRD) is T2DM. Extensive work is being done to delineate the pathogenesis of DN and to extend possible remedies. This review is intended to understand the nature of DN risk factors, progression, effects of glycemic levels, and stages of DN. We also explored the novel diagnostic and therapeutic approaches for DN such as gene therapy and stem cell treatments.
Collapse
Affiliation(s)
- Vijayakumar Natesan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar-608002, Tamilnadu, India
| | - Sung-Jin Kim
- Department of Pharmacology and Toxicology, Metabolic Diseases Research Laboratory, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
99
|
Diabetic kidney disease: new clinical and therapeutic issues. Joint position statement of the Italian Diabetes Society and the Italian Society of Nephrology on "The natural history of diabetic kidney disease and treatment of hyperglycemia in patients with type 2 diabetes and impaired renal function". J Nephrol 2021; 33:9-35. [PMID: 31576500 PMCID: PMC7007429 DOI: 10.1007/s40620-019-00650-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aims This joint document of the Italian Diabetes Society and the Italian Society of Nephrology reviews the natural history of diabetic kidney disease (DKD) in the light of the recent epidemiological literature and provides updated recommendations on anti-hyperglycemic treatment with non-insulin agents. Data Synthesis Recent epidemiological studies have disclosed a wide heterogeneity of DKD. In addition to the classical albuminuric phenotype, two new albuminuria-independent phenotypes have emerged, i.e., “nonalbuminuric renal impairment” and “progressive renal decline”, suggesting that DKD progression toward end-stage kidney disease (ESKD) may occur through two distinct pathways, albuminuric and nonalbuminuric. Several biomarkers have been associated with decline of estimated glomerular filtration rate (eGFR) independent of albuminuria and other clinical variables, thus possibly improving ESKD prediction. However, the pathogenesis and anatomical correlates of these phenotypes are still unclear. Also the management of hyperglycemia in patients with type 2 diabetes and impaired renal function has profoundly changed during the last two decades. New anti-hyperglycemic drugs, which do not cause hypoglycemia and weight gain and, in some cases, seem to provide cardiorenal protection, have become available for treatment of these individuals. In addition, the lowest eGFR safety thresholds for some of the old agents, particularly metformin and insulin secretagogues, have been reconsidered. Conclusions The heterogeneity in the clinical presentation and course of DKD has important implications for the diagnosis, prognosis, and possibly treatment of this complication. The therapeutic options for patients with type 2 diabetes and impaired renal function have substantially increased, thus allowing a better management of these individuals.
Collapse
|
100
|
Urinary biomarkers as point-of-care tests for predicting progressive deterioration of kidney function in congenital anomalies of kidney and urinary tract: trefoil family factors (TFFs) as the emerging biomarkers. Pediatr Nephrol 2021; 36:1465-1472. [PMID: 33420628 DOI: 10.1007/s00467-020-04841-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/02/2020] [Accepted: 10/22/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND Children with congenital anomalies of kidney and urinary tract (CAKUT) are at high risk of progressive deterioration of kidney function and further developing stage 5 chronic kidney disease (CKD 5), even after a successful surgery. This prospective study was designed to determine whether urinary biomarkers can predict progressive deterioration of kidney function in children with CAKUT. METHODS The study included 50 consecutive children, aged < 14 years, who were diagnosed with congenital uropathies (PUV, VUR, and PUJO) and 20 age-matched controls. Examination of four urinary biomarkers, i.e., trefoil family factors (TFF) 1 and 3, neutrophil gelatinase-associated lipocalin (NGAL) and microalbuminuria (MALB) was done at the beginning of follow-up. Kidney function was assessed, at the beginning and after 12-months of follow-up, by technetium-99m diethylene triamine pentaacetic acid (DTPA) and technetium-99m dimercaptosuccinic acid (DMSA) scans. Progressive deterioration in the kidney function was defined as a fall in the GFR from ≥ 60 to < 60 ml/min/1.73 m2 on comparing the baseline and latest DTPA scans; and/or new-onset cortical scar/scars or increase in the size of previous scar/scars on serial DMSA scans. Group 1 and group 2 included children without and with progressive functional deterioration respectively. RESULTS The median (IQR) age of children with CAKUT and controls was 3 (1.5-5) and 2.3 (1.2-3.6) years, respectively, and showed no significant difference (p = 0.29). Median concentrations of TFF1, TFF3, NGAL, and microalbumin in patients were 44.5, 176.5, 281.2, and 15.5 mcg/gCr, respectively, and were significantly elevated as compared to controls (p < 0.05). Children belonging to group 2 had significantly higher concentration of biomarkers as compared to those in group 1. TFF3 was found have the highest AUC (0.9198) on ROC curve for predicting progressive functional deterioration. CONCLUSION Urinary TFFs, NGAL, and microalbumin significantly correlate with progressive deterioration of kidney function in children harboring CAKUT. TFF3, with the strongest prediction of functional deterioration, is an emerging peptide showing sufficient potential to be included in the biomarker panel. Graphical abstract.
Collapse
|