1
|
Miloševski-Lomić G, Kotur-Stevuljević J, Paripović D, Nikolovski S, Lazić J, Rodić P, Milošević G, Mitrović J, Vukmir B, Petrović A, Peco-Antić A. Urinary biomarkers in prediction of subclinical acute kidney injury in pediatric oncology patients treated with nephrotoxic agents. BMC Nephrol 2025; 26:159. [PMID: 40148769 PMCID: PMC11951557 DOI: 10.1186/s12882-025-04085-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication in pediatric oncology patients, most often caused by nephrotoxic drugs. We aimed to assess whether levels of urinary kidney injury molecule-1 (uKIM-1), neutrophil gelatinase-associated lipocalin (uNGAL), liver fatty acid binding protein (uL-FABP) and Vanin-1 (uVNN-1), individually and in combination-integrated could be early markers for cytotoxic treatment induced AKI. METHODS Children with different malignant diseases treated with cisplatin (CIS) or ifosfamide (IFO) were included. AKI was defined using pediatric KDIGO (Kidney Disease Improving Global Outcomes) criteria by comparing pretreatment serum creatinine (sCr) values with those acquired at 48 h after the first or second chemotherapy cycle. Five serum (at baseline, 2, 6, 24 and 48 h after treatment) and four urine samples (at baseline, 2, 6 and 24 h after treatment) were obtained. Urinary biomarkers (uBm) were normalized to urine creatinine. RESULTS Thirty-eight patients were assessed. Within 48 h following chemotherapy 6 (15.79%) patients experienced AKI. Patients with AKI were younger and tend to have lower baseline sCr values than patients without AKI, but these differences were not statistically significant. Compared to baselines, all uBm were significantly increased during the first 6 h while sCr concentrations did not change significantly during the study period. The median increases in uBm during the first 6 h after treatment were 529.8% (interquartile range - IQR, 63.9-1835.2%) - 2194.0% (IQR, 255.3-4695.5%) in AKI vs. 302.2% (IQR 114.6-561.2%) -429.8% (156.5-1467.0%) in non-AKI group depending of tested uBm. The magnitude of these changes over time didn't differ significantly between groups. The area under receiver operator curve (AUC) for uL-FABP and uNGAL at 24 h after chemotherapy were 0.81 and 0.72, respectively. The ROC analysis revealed that the other individual biomarkers' performance at any time-point wasn't statistically significant (AUC < 0.7). A model of integrated-combined uBm, 2 h (AUC 0.78), 6 h (AUC 0.85) and 24 h after (AUC 0.92) treatment with CIS and/or IFO showed good utility for early AKI prediction. CONCLUSIONS The results of this study support that the use of the uBm to improves early AKI prediction in patients receiving CIS and/or IFO containing chemotherapy. Further studies on larger comparable groups of patients are needed.
Collapse
Affiliation(s)
- Gordana Miloševski-Lomić
- Faculty of Medicine, University of Belgrade, Belgrade, 11000, Serbia.
- Nephrology Department, University Children's Hospital, Tiršova 10, Belgrade, 11000, Serbia.
| | - Jelena Kotur-Stevuljević
- Department for Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, 11000, Serbia
| | - Dušan Paripović
- Faculty of Medicine, University of Belgrade, Belgrade, 11000, Serbia
- Nephrology Department, University Children's Hospital, Tiršova 10, Belgrade, 11000, Serbia
| | - Srdjan Nikolovski
- Nephrology Department, University Children's Hospital, Tiršova 10, Belgrade, 11000, Serbia
- Loyola University Chicago Medical Center, Maywood, IL, 60153, USA
| | - Jelena Lazić
- Faculty of Medicine, University of Belgrade, Belgrade, 11000, Serbia
- Hematology and Oncology Department, University Children's Hospital, Belgrade, 11000, Serbia
| | - Predrag Rodić
- Faculty of Medicine, University of Belgrade, Belgrade, 11000, Serbia
- Hematology and Oncology Department, University Children's Hospital, Belgrade, 11000, Serbia
| | - Goran Milošević
- Faculty of Medicine, University of Belgrade, Belgrade, 11000, Serbia
- Hematology and Oncology Department, University Children's Hospital, Belgrade, 11000, Serbia
| | - Jadranka Mitrović
- Biochemistry Department, University Children's Hospital, Belgrade, 11000, Serbia
| | - Biljana Vukmir
- Biochemistry Department, University Children's Hospital, Belgrade, 11000, Serbia
| | - Ana Petrović
- Nephrology Department, University Children's Hospital, Tiršova 10, Belgrade, 11000, Serbia
| | - Amira Peco-Antić
- Faculty of Medicine, University of Belgrade, Belgrade, 11000, Serbia
- Nephrology Department, University Children's Hospital, Tiršova 10, Belgrade, 11000, Serbia
| |
Collapse
|
2
|
Rashed HAEH, El-Shenawy NS, El-Fahla NA. Influences of parasitic stress on the health condition of African Catfish (Clarias gariepinus): Biochemical and histopathological alterations. Mol Biochem Parasitol 2025; 262:111677. [PMID: 40064449 DOI: 10.1016/j.molbiopara.2025.111677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/15/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
This study investigates the prevalence, severity, and impacts of parasitic infestations in Clarias gariepinus. Additionally, the study assesses the detrimental impacts of parasite infestation on the health condition of affected catfish, focusing on biochemical and histopathological alterations. A total of 160 fish were sampled from local markets. Parasitological examinations involved the dissection of key organs from each fish. The organs were processed and examined microscopically for parasites identified based on morphometric characteristics. Parasitological indices such as prevalence, mean intensity, and abundance were calculated. Fish blood and liver samples were collected to assess hematological and biochemical parameters. Microscopic and ultrastructural examinations identified the gills and liver as highly infected organs, so they were utilized for transmission electron microscopy (TEM). Analysis of catfish tissues unveiled the existence of Cyathocotylid sp. and Prohemistomum vivax, across all organs with dominance noted in the liver, emphasizing their pathogenic significance and notable ability to invade and establish within multiple organs or the immunocompromised response of the host. Meanwhile, Centrocestus formosanus and Quadriacanthus aegyptiacus were exclusively detected in the gills, with an overall parasitic infection rate of 60 %. The present study is one of the few studies documenting Centrocestus sp. in catfish which reflects its ability to spread in new hosts and environments. A novel morphological dimension was recorded for the recovered metacercariae. The hematological, along with the identified lesions from light histological and TEM examinations in heavily infected catfish, indicate the detrimental impact of parasite infiltration on fish health status. Besides the biochemical biomarkers were significantly (p ≤ 0.05) affected by increasing the degree of infection. This study underscores the profound influence of parasitic infestations on the health of C. gariepinus, emphasizing the urgent need for effective management strategies in aquaculture to mitigate these effects, the spread of new pathogens, and ensure the sustainability and productivity of catfish farming. By integrating parasitological, morphological, histopathological, and biochemical analyses, this research provides valuable insights that contribute to better health management strategies in aquaculture and a deeper understanding of parasite biology.
Collapse
Affiliation(s)
| | - Nahla S El-Shenawy
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt.
| | - Nadia A El-Fahla
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
3
|
Mistry K, Sadarangani S, Moreno D, Mejia SM, Moledina DG, Sise ME. Novel Biomarkers and Imaging Tests for Acute Kidney Injury Diagnosis in Patients with Cancer. KIDNEY360 2025; 6:167-174. [PMID: 39575585 PMCID: PMC11793192 DOI: 10.34067/kid.0000000660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025]
Abstract
The lack of noninvasive urine and blood-based biomarkers for the diagnosis of AKI in patients with cancer is an area of significant unmet clinical need. Traditional noninvasive diagnostic tools that are currently used in the clinic, such as creatinine and cystatin C-based eGFR measurements, urinalysis, urine sediment examination, urine protein quantification, and urine electrolyte measurement, lack the sensitivity and specificity to distinguish between the various underlying etiologies of AKI in patients with cancer. Imaging-based diagnostics can be helpful to rule out urinary obstruction, but also lack sensitivity and specificity to diagnose the etiology of AKI. Kidney biopsy is often required for definitive diagnosis. As our scientific understanding of the biological pathways that are dysregulated in AKI has advanced, there has been considerable interest in developing new biomarkers for AKI. For example, the diagnosis of acute interstitial nephritis, which can occur in patients treated with immune checkpoint inhibitors, promises to be revolutionized by the incorporation of urinary testing for inflammatory biomarkers, such as C-X-C motif ligand 9, TNF- α , and IL-9. In the case of cisplatin administration, biomarkers such as neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 may improve prognostication, differentiating between persistent AKI resulting from acute tubular injury versus prerenal azotemia. The development and validation of blood, urine, and imaging biomarkers into widely used diagnostic tests will require a concerted effort, but could improve diagnosis, management, and prognostication for a growing group of patients who are at high risk of developing AKI during the course of their illness.
Collapse
Affiliation(s)
- Kavita Mistry
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Sagar Sadarangani
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Daiana Moreno
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Sherley M. Mejia
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Dennis G. Moledina
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Internal Medicine, Clinical and Translational Research Accelerator, New Haven, Connecticut
| | - Meghan E. Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
4
|
Chang J, Pais GM, Barreto EF, Young B, Scott H, Schwartz Z, Cartwright C, Jubrail R, Srivastava A, Scheetz MH. Past, present, and future biomarkers of kidney function and injury: The relationship with antibiotics. Int J Antimicrob Agents 2024; 64:107332. [PMID: 39245327 DOI: 10.1016/j.ijantimicag.2024.107332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
Routinely used kidney biomarkers of injury and function such as serum creatinine and urine albumin to creatinine ratio, are neither sensitive nor specific. Future biomarkers are being developed for clinical use and have already been included in guidance from groups such as the U.S. Food and Drug Administration and the Predictive Safety Testing Consortium. These biomarkers have important implications for early identification of kidney injury and more accurate measurement of kidney function. Many antibiotics are either eliminated by the kidney or can cause clinically significant nephrotoxicity. As a result, clinicians should be familiar with new biomarkers of kidney function and injury, their place in clinical practice, and applications for antibiotic dosing.
Collapse
Affiliation(s)
- Jack Chang
- Department of Pharmacy Practice (J.C., G.M.P., R.J., M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA; Pharmacometrics Center of Excellence (J.C., G.M.P., M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA; Department of Pharmacy (J.C., M.H.S.), Northwestern Memorial Hospital, Chicago, Illinois, USA
| | - Gwendolyn M Pais
- Department of Pharmacy Practice (J.C., G.M.P., R.J., M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA; Pharmacometrics Center of Excellence (J.C., G.M.P., M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA
| | - Erin F Barreto
- Department of Pharmacy (E.F.B.), Mayo Clinic, Rochester, Minnesota, USA
| | - Bryce Young
- Chicago College of Osteopathic Medicine (B.Y., H.S., Z.S., C.C.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA
| | - Haley Scott
- Chicago College of Osteopathic Medicine (B.Y., H.S., Z.S., C.C.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA
| | - Zachary Schwartz
- Chicago College of Osteopathic Medicine (B.Y., H.S., Z.S., C.C.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA
| | - Collin Cartwright
- Chicago College of Osteopathic Medicine (B.Y., H.S., Z.S., C.C.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA
| | - Raymond Jubrail
- Department of Pharmacy Practice (J.C., G.M.P., R.J., M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA
| | - Anand Srivastava
- Division of Nephrology (A.S.), University of Illinois-Chicago, Chicago, Illinois, USA
| | - Marc H Scheetz
- Department of Pharmacy Practice (J.C., G.M.P., R.J., M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA; Pharmacometrics Center of Excellence (J.C., G.M.P., M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA; Department of Pharmacy (J.C., M.H.S.), Northwestern Memorial Hospital, Chicago, Illinois, USA; Department of Pharmacology (M.H.S.), Midwestern University-Downers Grove Campus, Downers Grove, Illinois, USA.
| |
Collapse
|
5
|
Gao J, Chen CY, Tu J, Geng HY, Li HR, Sun JS, Wang NN, Huang YL. [Early identification of acute kidney injury in children with primary nephrotic syndrome]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:921-925. [PMID: 39267506 PMCID: PMC11404463 DOI: 10.7499/j.issn.1008-8830.2405033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
OBJECTIVES To investigate the incidence and risk factors for acute kidney injury (AKI) in children with primary nephrotic syndrome (PNS), as well as the role of neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) in the early identification of AKI in these children. METHODS A prospective collection of clinical data from children hospitalized with PNS at the Children's Hospital of the Capital Institute of Pediatrics from January 2021 to October 2022 was conducted. The children were divided into two groups based on the presence of AKI: the AKI group (47 cases) and the non-AKI group (169 cases). The risk factors for AKI in children with PNS were identified by multivariate logistic regression analysis. Urinary KIM-1 and NGAL levels were compared between the AKI and non-AKI groups, as well as among the different stages of AKI. RESULTS The incidence of AKI in children with PNS was 21.8%. Multivariate logistic regression analysis revealed that steroid-resistant nephrotic syndrome, gastrointestinal infections, and heavy proteinuria were independent risk factors for AKI in these children with PNS (P<0.05). Urinary KIM-1 and NGAL levels were higher in the AKI group compared to the non-AKI group (P<0.05), and the urinary NGAL and KIM-1 levels in the AKI stage 2 and stage 3 subgroups were higher than those in the AKI stage 1 subgroup (P<0.017). CONCLUSIONS KIM-1 and NGAL can serve as biomarkers for the early diagnosis of AKI in children with PNS. Identifying high-risk populations for AKI in children with PNS and strengthening the monitoring of related risk factors is of significant importance.
Collapse
Affiliation(s)
- Jie Gao
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Chao-Ying Chen
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Juan Tu
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Hai-Yun Geng
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Hua-Rong Li
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Jin-Shan Sun
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Nan-Nan Wang
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Yong-Li Huang
- Department of Kidney Internal Medicine, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| |
Collapse
|
6
|
Shirazi M, Cianfarini C, Ismail A, Wysocki J, Wang JJ, Ye M, Zhang ZJ, Batlle D. Altered kidney distribution and loss of ACE2 into the urine in acute kidney injury. Am J Physiol Renal Physiol 2024; 327:F412-F425. [PMID: 38961845 PMCID: PMC11460339 DOI: 10.1152/ajprenal.00237.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
There are diverse pathophysiological mechanisms involved in acute kidney injury (AKI). Among them, overactivity of the renin-angiotensin system (RAS) has been described. Angiotensin-converting enzyme 2 (ACE2) is a tissue RAS enzyme expressed in the apical border of proximal tubules. Given the important role of ACE2 in the metabolism of angiotensin II, this study aimed to characterize kidney and urinary ACE2 in a mouse model of AKI. Ischemia-reperfusion injury (IRI) was induced in C57BL/6 mice by clamping of the left renal artery followed by removal of the right kidney. In kidneys harvested 48 h after IRI, immunostaining revealed a striking maldistribution of ACE2 including spillage into the tubular lumen and the presence of ACE2-positive luminal casts in the medulla. In cortical membranes, ACE2 protein and enzymatic activity were both markedly reduced (37 ± 4 vs. 100 ± 6 ACE2/β-actin, P = 0.0004, and 96 ± 14 vs. 152 ± 6 RFU/μg protein/h, P = 0.006). In urine, full-length membrane-bound ACE2 protein (100 kDa) was markedly increased (1,120 ± 405 vs. 100 ± 46 ACE2/µg creatinine, P = 0.04), and casts stained for ACE2 were recovered in the urine sediment. In conclusion, in AKI caused by IRI, there is a marked loss of ACE2 from the apical tubular border with deposition of ACE2-positive material in the medulla and increased urinary excretion of full-length membrane-bound ACE2 protein. The deficiency of tubular ACE2 in AKI suggests that provision of this enzyme could have therapeutic applications and that its excretion in the urine may also serve as a diagnostic marker of severe proximal tubular injury.NEW & NOTEWORTHY This study provides novel insights into the distribution of kidney ACE2 in a model of AKI by IRI showing a striking detachment of apical ACE2 from proximal tubules and its loss in urine and urine sediment. The observed deficiency of kidney ACE2 protein and enzymatic activity in severe AKI suggests that administration of forms of this enzyme may mitigate AKI and that urinary ACE2 may serve as a potential biomarker for tubular injury.
Collapse
Affiliation(s)
- Mina Shirazi
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Cosimo Cianfarini
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ahmed Ismail
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Jan Wysocki
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Jiao-Jing Wang
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Minghao Ye
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Zheng Jenny Zhang
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Daniel Batlle
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
7
|
Arndt T, Keresztes M, Olivier B, Boone L, Chanut F, Ennulat D, Evans E, Freyberger A, Johannes S, Kuper CF, Maliver P, O'Brien P, Ramaiah L, Roman I, Strauss V, Vinken P, Walker D, Winter M, Pohlmeyer-Esch G, Tomlinson L. Considerations for the Identification and Conveyance of Clinical Pathology Findings in Preclinical Toxicity Studies: Results From the 9th ESTP International Expert Workshop. Toxicol Pathol 2024; 52:319-332. [PMID: 38661116 DOI: 10.1177/01926233241245108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The European Society of Toxicologic Pathology (ESTP) organized a panel of 24 international experts from many fields of toxicologic clinical pathology (e.g., industry, academia, and regulatory) that came together in 2021 to align the use of terminology to convey the importance of clinical pathology findings in preclinical toxicity studies. An additional goal consisted of how to identify important findings in standard and nonstandard clinical pathology associated endpoints. This manuscript summarizes the information and opinions discussed and shared at the ninth ESTP International Expert Workshop, April 5 to 6, 2022. In addition to terminology usage, the workshop considered topics related to the identification and conveyance of the importance of test item-related findings. These topics included sources of variability, comparators, statistics, reporting, correlations to other study data, nonstandard biomarkers, indirect/secondary findings, and an overall weight-of-evidence approach.
Collapse
Affiliation(s)
- Tara Arndt
- Altasciences Preclinical, Laval, Quebec, Canada
- Altasciences Preclinical, Seattle, Washington, USA
| | | | | | - L Boone
- Labcorp, Madison, Wisconsin, USA
| | | | - D Ennulat
- GlaxoSmithKline (Retired), King of Prussia, Pennsylvania, USA
| | - Ellen Evans
- Pfizer (Retired), Waterford, Connecticut, USA
| | | | | | | | - Pierre Maliver
- Roche Pharma Research and Early Development, Basel, Switzerland
| | | | - Lila Ramaiah
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Ian Roman
- GlaxoSmithKline, Ware, United Kingdom
| | | | | | - Dana Walker
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Michael Winter
- Roche Pharma Research and Early Development, Basel, Switzerland
| | | | | |
Collapse
|
8
|
Lisa A, Carbone F, Liberale L, Montecucco F. The Need to Identify Novel Markers for Early Renal Injury in Cardiorenal Syndrome. Cells 2024; 13:1283. [PMID: 39120314 PMCID: PMC11311518 DOI: 10.3390/cells13151283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
The term "Cardiorenal Syndrome" (CRS) refers to the complex interplay between heart and kidney dysfunction. First described by Robert Bright in 1836, CRS was brought to its modern view by Ronco et al. in 2008, who defined it as one organ's primary dysfunction leading to secondary dysfunction in the other, a view that led to the distinction of five different types depending on the organ of primary dysfunction and the temporal pattern (acute vs. chronic). Their pathophysiology is intricate, involving various hemodynamic, neurohormonal, and inflammatory processes that result in damage to both organs. While traditional biomarkers have been utilized for diagnosing and prognosticating CRS, they are inadequate for the early detection of acute renal damage. Hence, there is a pressing need to discover new biomarkers to enhance clinical outcomes and treatment approaches.
Collapse
Affiliation(s)
- Anna Lisa
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy (F.C.); (L.L.)
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy (F.C.); (L.L.)
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy (F.C.); (L.L.)
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy (F.C.); (L.L.)
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy
| |
Collapse
|
9
|
Zhang D, Li L, Huang W, Hu C, Zhu W, Hu B, Li J. Vasoactive-Inotropic Score as a Promising Predictor of Acute Kidney Injury in Adult Patients Requiring Extracorporeal Membrane Oxygenation. ASAIO J 2024; 70:586-593. [PMID: 38324707 PMCID: PMC11210947 DOI: 10.1097/mat.0000000000002158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Acute kidney injury (AKI) is a common complication in patients supported by extracorporeal membrane oxygenation (ECMO). Vasoactive-Inotropic Score (VIS) serves as an indicator of the extent of cardiovascular drug support provided. Our objective is to assess the relationship between the VIS and ECMO-associated AKI (EAKI). This single-center retrospective study extracted adult patients treated with ECMO between August 2016 and September 2022 from an intensive care unit (ICU) in a university hospital. A total of 126 patients requiring ECMO support were included in the study, of which 76% developed AKI. Multivariate logistic regression analysis identified VIS-max Day1 (odds ratio [OR]: 1.025, 95% confidence interval [CI]: 1.007-1.044, p = 0.006), VIS-max Day2 (OR: 1.038, 95% CI: 1.007-1.069, p = 0.015), VIS-mean Day1 (OR: 1.048, 95% CI: 1.013-1.084, p = 0.007), and VIS-mean Day2 (OR: 1.059, 95% CI: 1.014-1.107, p = 0.010) as independent risk factors for EAKI. VIS-max Day1 showing the best predictive effect (Area under the receiver operating characteristic curve (AUROC): 0.80, sensitivity: 71.87%, specificity: 80.00%) for EAKI with a cutoff value of 33.33. Surprisingly, VIS-mean Day2 was also excellent at predicting 7 day mortality (AUROC: 0.77, sensitivity: 87.50%, specificity: 56.38%) with a cutoff value of 8.67. In conclusion, VIS could independently predict EAKI and 7 day mortality in patients with ECMO implantation, which may help clinicians to recognize the poor prognosis in time for early intervention.
Collapse
Affiliation(s)
- Dandan Zhang
- From the Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Lu Li
- From the Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Weipeng Huang
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chang Hu
- From the Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Weiwei Zhu
- From the Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Bo Hu
- From the Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jianguo Li
- From the Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| |
Collapse
|
10
|
Tutunea-Fatan E, Arumugarajah S, Suri RS, Edgar CR, Hon I, Dikeakos JD, Gunaratnam L. Sensing Dying Cells in Health and Disease: The Importance of Kidney Injury Molecule-1. J Am Soc Nephrol 2024; 35:795-808. [PMID: 38353655 PMCID: PMC11164124 DOI: 10.1681/asn.0000000000000334] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
Abstract
Kidney injury molecule-1 (KIM-1), also known as T-cell Ig and mucin domain-1 (TIM-1), is a widely recognized biomarker for AKI, but its biological function is less appreciated. KIM-1/TIM-1 belongs to the T-cell Ig and mucin domain family of conserved transmembrane proteins, which bear the characteristic six-cysteine Ig-like variable domain. The latter enables binding of KIM-1/TIM-1 to its natural ligand, phosphatidylserine, expressed on the surface of apoptotic cells and necrotic cells. KIM-1/TIM-1 is expressed in a variety of tissues and plays fundamental roles in regulating sterile inflammation and adaptive immune responses. In the kidney, KIM-1 is upregulated on injured renal proximal tubule cells, which transforms them into phagocytes for clearance of dying cells and helps to dampen sterile inflammation. TIM-1, expressed in T cells, B cells, and natural killer T cells, is essential for cell activation and immune regulatory functions in the host. Functional polymorphisms in the gene for KIM-1/TIM-1, HAVCR1 , have been associated with susceptibility to immunoinflammatory conditions and hepatitis A virus-induced liver failure, which is thought to be due to a differential ability of KIM-1/TIM-1 variants to bind phosphatidylserine. This review will summarize the role of KIM-1/TIM-1 in health and disease and its potential clinical applications as a biomarker and therapeutic target in humans.
Collapse
Affiliation(s)
- Elena Tutunea-Fatan
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
| | - Shabitha Arumugarajah
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rita S. Suri
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Division of Nephrology, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cassandra R. Edgar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Ingrid Hon
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Lakshman Gunaratnam
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
11
|
Ono K, Maeshima A, Nagayama I, Kubo T, Yagisawa T, Nagata D. Urinary Epidermal Growth Factor Level as a Noninvasive Indicator of Tubular Repair in Patients with Acute Kidney Injury. Diagnostics (Basel) 2024; 14:947. [PMID: 38732362 PMCID: PMC11083164 DOI: 10.3390/diagnostics14090947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/28/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Epidermal growth factor (EGF), an essential factor for the proliferation and survival of renal tubular cells, is expressed by distal tubules and normally excreted via urine. Previous studies in rats demonstrated that acute tubular injury reduces urinary EGF levels. However, it is unclear whether urinary EGF is a suitable monitoring marker of tubular repair status after acute kidney injury (AKI) in humans. To address this question, we measured serum and urinary EGF in patients with AKI (n = 99) using ELISA and investigated whether urinary EGF levels were associated with the severity of tubular injury and renal prognosis. Urinary EGF was abundant in healthy controls but showed a significant decrease in AKI patients (14,522 ± 2190 pg/mL vs. 3201 ± 459.7 pg/mL, p < 0.05). The urinary EGF level in patients with renal AKI was notably lower than that in patients with pre-renal AKI. Furthermore, the urinary EGF level in patients with AKI stage 3 was significantly lower than that in patients with AKI stage 1. Urinary EGF levels were negatively correlated with urinary β-2MG and serum creatinine levels but positively correlated with hemoglobin levels and eGFR. Urinary EGF was not significantly correlated with urinary NAG, α-1MG, L-FABP, NGAL, KIM-1, or urinary protein concentrations. No significant correlation was observed between serum and urinary EGF levels, suggesting that urinary EGF is derived from the renal tubules rather than the blood. In living renal transplantation donors, the urinary EGF/Cr ratio was approximately half the preoperative urinary EGF/Cr ratio after unilateral nephrectomy. Collectively, these data suggest that urinary EGF is a suitable noninvasive indicator of not only the volume of functional normal renal tubules but also the status of tubular repair after AKI.
Collapse
Affiliation(s)
- Kazutoshi Ono
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Akito Maeshima
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kawagoe 350-8550, Japan
| | - Izumi Nagayama
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Kawagoe 350-8550, Japan
| | - Taro Kubo
- Department of Renal Surgery and Transplantation, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Takashi Yagisawa
- Department of Renal Surgery and Transplantation, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan
| |
Collapse
|
12
|
Brown T, Defarges A, Monteith G, Appleby R, Bienzle D. Determination of the reference interval for urine kidney injury molecule-1 in 50 healthy cats. J Feline Med Surg 2024; 26:1098612X241238923. [PMID: 38647460 PMCID: PMC11103318 DOI: 10.1177/1098612x241238923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
OBJECTIVES The aim of the present study was to establish a reference interval (RI) for urine kidney injury molecule-1 (KIM-1) in healthy cats. METHODS History, physical examination, blood pressure, and feline immunodeficiency virus and feline leukemia virus serology status were determined. A complete blood cell count, serum biochemical profile, urinalysis and kidney ultrasound were performed, and N-terminal pro-brain natriuretic peptide, total thyroxine (TT4) and urine KIM-1 were measured. An RI was calculated and the effect of age, sex, body condition score (BCS), blood pressure, symmetric dimethylarginine (SDMA), serum creatinine concentration (SCr), phosphorus, TT4, urine specific gravity (USG) and mid-sagittal kidney length on urine KIM-1 was evaluated using a general linear model. RESULTS Of 69 recruited cats, 50 met the inclusion criteria. There were 35 male cats and 15 female cats, with a median age of 4.3 years (range 1.0-12.3), median weight of 5.11 kg (range 2.52-8.45) and median BCS of 6/9 (range 3-8). The median serum concentrations were SDMA 11.0 µg/dl (range 2-14), SCr 88.5 µmol/l (range 47-136), phosphorus 1.41 mmol/l (range 0.8-2.2) and TT4 32.0 nmol/l (range 17-51). Median USG was 1.057 (range 1.035-1.076), mid-sagittal left kidney length was 3.50 cm (range 2.94-4.45) and mid-sagittal right kidney length was 3.70 cm (range 3.06-4.55). The derived RI for urine KIM-1 was 0.02-0.68. USG was a significant (P <0.001) predictor of urine KIM-1. Individually, age, sex, blood pressure, BCS, SDMA, SCr, phosphorus, TT4 and mid-sagittal kidney length were not significant predictors of urine KIM-1. In a multivariate model, if combined with USG, SDMA concentration was predictive (P = 0.030) of urine KIM-1. CONCLUSIONS AND RELEVANCE Urine concentration was significantly correlated with urine KIM-1, which will be an important consideration when interpreting findings in cats with potential kidney injury.
Collapse
Affiliation(s)
- Tori Brown
- Mississauga Oakville Veterinary Emergency Hospital, Oakville, ON, Canada
| | - Alice Defarges
- Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| | - Gabrielle Monteith
- Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| | - Ryan Appleby
- Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| | | |
Collapse
|
13
|
Haykir B, Moser SO, Pastor-Arroyo EM, Schnitzbauer U, Radvanyi Z, Prucker I, Qiu D, Fiedler D, Saiardi A, Jessen HJ, Hernando N, Wagner CA. The Ip6k1 and Ip6k2 Kinases Are Critical for Normal Renal Tubular Function. J Am Soc Nephrol 2024; 35:441-455. [PMID: 38317282 PMCID: PMC11000740 DOI: 10.1681/asn.0000000000000303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/24/2023] [Indexed: 02/07/2024] Open
Abstract
SIGNIFICANCE STATEMENT Kidneys are gatekeepers of systemic inorganic phosphate balance because they control urinary phosphate excretion. In yeast and plants, inositol hexakisphosphate kinases (IP6Ks) are central to regulate phosphate metabolism, whereas their role in mammalian phosphate homeostasis is mostly unknown. We demonstrate in a renal cell line and in mice that Ip6k1 and Ip6k2 are critical for normal expression and function of the major renal Na + /Pi transporters NaPi-IIa and NaPi-IIc. Moreover, Ip6k1/2-/- mice also show symptoms of more generalized kidney dysfunction. Thus, our results suggest that IP6Ks are essential for phosphate metabolism and proper kidney function in mammals. BACKGROUND Inorganic phosphate is an essential mineral, and its plasma levels are tightly regulated. In mammals, kidneys are critical for maintaining phosphate homeostasis through mechanisms that ultimately regulate the expression of the Na + /Pi cotransporters NaPi-IIa and NaPi-IIc in proximal tubules. Inositol pyrophosphate 5-IP 7 , generated by IP6Ks, is a main regulator of phosphate metabolism in yeast and plants. IP6Ks are conserved in mammals, but their role in phosphate metabolism in vivo remains unexplored. METHODS We used in vitro (opossum kidney cells) and in vivo (renal tubular-specific Ip6k1/2-/- mice) models to analyze the role of IP6K1/2 in phosphate homeostasis in mammals. RESULTS In both systems, Ip6k1 and Ip6k2 are responsible for synthesis of 5-IP 7 . Depletion of Ip6k1/2 in vitro reduced phosphate transport and mRNA expression of Na + /Pi cotransporters, and it blunts phosphate transport adaptation to changes in ambient phosphate. Renal ablation of both kinases in mice also downregulates the expression of NaPi-IIa and NaPi-IIc and lowered the uptake of phosphate into proximal renal brush border membranes. In addition, the absence of Ip6k1 and Ip6k2 reduced the plasma concentration of fibroblast growth factor 23 and increased bone resorption, despite of which homozygous males develop hypophosphatemia. Ip6k1/2-/- mice also show increased diuresis, albuminuria, and hypercalciuria, although the morphology of glomeruli and proximal brush border membrane seemed unaffected. CONCLUSIONS Depletion of renal Ip6k1/2 in mice not only altered phosphate homeostasis but also dysregulated other kidney functions.
Collapse
Affiliation(s)
- Betül Haykir
- Switzerland and National Center of Competence in Research NCCR Kidney.CH, Institute of Physiology, University of Zurich, Zürich, Switzerland
| | - Seraina Olivia Moser
- Switzerland and National Center of Competence in Research NCCR Kidney.CH, Institute of Physiology, University of Zurich, Zürich, Switzerland
| | - Eva Maria Pastor-Arroyo
- Switzerland and National Center of Competence in Research NCCR Kidney.CH, Institute of Physiology, University of Zurich, Zürich, Switzerland
| | - Udo Schnitzbauer
- Switzerland and National Center of Competence in Research NCCR Kidney.CH, Institute of Physiology, University of Zurich, Zürich, Switzerland
| | - Zsuzsa Radvanyi
- Switzerland and National Center of Competence in Research NCCR Kidney.CH, Institute of Physiology, University of Zurich, Zürich, Switzerland
| | - Isabel Prucker
- The Center for Integrative Biological Signalling Studies, Institute of Organic Chemistry and CIBSS, University of Freiburg, Freiburg, Germany
| | - Danye Qiu
- The Center for Integrative Biological Signalling Studies, Institute of Organic Chemistry and CIBSS, University of Freiburg, Freiburg, Germany
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Adolfo Saiardi
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Henning J. Jessen
- The Center for Integrative Biological Signalling Studies, Institute of Organic Chemistry and CIBSS, University of Freiburg, Freiburg, Germany
| | - Nati Hernando
- Switzerland and National Center of Competence in Research NCCR Kidney.CH, Institute of Physiology, University of Zurich, Zürich, Switzerland
| | - Carsten A. Wagner
- Switzerland and National Center of Competence in Research NCCR Kidney.CH, Institute of Physiology, University of Zurich, Zürich, Switzerland
| |
Collapse
|
14
|
Deng JW, Li CY, Huang YP, Liu WF, Zhang Q, Long J, Wu WQ, Huang LH, Zeng GH, Sun XY. Mechanism of Porphyra Yezoensis Polysaccharides in Inhibiting Hyperoxalate-Induced Renal Injury and Crystal Deposition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6372-6388. [PMID: 38471112 DOI: 10.1021/acs.jafc.3c09152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Oxidative damage to the kidneys is a primary factor in the occurrence of kidney stones. This study explores the inhibitory effect of Porphyra yezoensis polysaccharides (PYP) on oxalate-induced renal injury by detecting levels of oxidative damage, expression of adhesion molecules, and damage to intracellular organelles and revealed the molecular mechanism by molecular biology methods. Additionally, we validated the role of PYP in vivo using a crystallization model of hyperoxalate-induced rats. PYP effectively scavenged the overproduction of reactive oxygen species (ROS) in HK-2 cells, inhibited the adhesion of calcium oxalate (CaOx) crystals on the cell surface, unblocked the cell cycle, restored the depolarization of the mitochondrial membrane potential, and inhibited cell death. PYP upregulated the expression of antioxidant proteins, including Nrf2, HO-1, SOD, and CAT, while decreasing the expression of Keap-1, thereby activating the Keap1/Nrf2 signaling pathway. PYP inhibited CaOx deposition in renal tubules in the rat crystallization model, significantly reduced high oxalate-induced renal injury, decreased the levels of the cell surface adhesion proteins, improved renal function in rats, and ultimately inhibited the formation of kidney stones. Therefore, PYP, which has crystallization inhibition and antioxidant properties, may be a therapeutic option for the treatment of kidney stones.
Collapse
Affiliation(s)
- Ji-Wang Deng
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Chun-Yao Li
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Ya-Peng Huang
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Wei-Feng Liu
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Quan Zhang
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Jun Long
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Wen-Qi Wu
- Department of Urology, Guangdong Key Laboratory of Urology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Ling-Hong Huang
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Guo-Hua Zeng
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Xin-Yuan Sun
- Department of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| |
Collapse
|
15
|
Singh R, Watchorn JC, Zarbock A, Forni LG. Prognostic Biomarkers and AKI: Potential to Enhance the Identification of Post-Operative Patients at Risk of Loss of Renal Function. Res Rep Urol 2024; 16:65-78. [PMID: 38476861 PMCID: PMC10928916 DOI: 10.2147/rru.s385856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Acute kidney injury (AKI) is a common complication after surgery and the more complex the surgery, the greater the risk. During surgery, patients are exposed to a combination of factors all of which are associated with the development of AKI. These include hypotension and hypovolaemia, sepsis, systemic inflammation, the use of nephrotoxic agents, tissue injury, the infusion of blood or blood products, ischaemia, oxidative stress and reperfusion injury. Given the risks of AKI, it would seem logical to conclude that early identification of patients at risk of AKI would translate into benefit. The conventional markers of AKI, namely serum creatinine and urine output are the mainstay of defining chronic kidney disease but are less suited to the acute phase. Such concerns are compounded in surgical patients given they often have significantly reduced mobility, suboptimal levels of nutrition and reduced muscle bulk. Many patients may also have misleadingly low serum creatinine and high urine output due to aggressive fluid resuscitation, particularly in intensive care units. Over the last two decades, considerable information has accrued with regard to the performance of what was termed "novel" biomarkers of AKI, and here, we discuss the most examined molecules and performance in surgical settings. We also discuss the application of biomarkers to guide patients' postoperative care.
Collapse
Affiliation(s)
- Rishabh Singh
- Department of Surgery, Royal Surrey Hospital, Guildford, Surrey, UK
| | - James C Watchorn
- Intensive Care Unit, Royal Berkshire NHS Foundation Trust, Reading, Berkshire, UK
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Lui G Forni
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surrey, UK
- School of Medicine, Kate Granger Building, University of Surrey, Guildford, UK
| |
Collapse
|
16
|
Burton JB, Silva-Barbosa A, Bons J, Rose J, Pfister K, Simona F, Gandhi T, Reiter L, Bernhardt O, Hunter CL, Goetzman ES, Sims-Lucas S, Schilling B. Substantial downregulation of mitochondrial and peroxisomal proteins during acute kidney injury revealed by data-independent acquisition proteomics. Proteomics 2024; 24:e2300162. [PMID: 37775337 DOI: 10.1002/pmic.202300162] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 10/01/2023]
Abstract
Acute kidney injury (AKI) manifests as a major health concern, particularly for the elderly. Understanding AKI-related proteome changes is critical for prevention and development of novel therapeutics to recover kidney function and to mitigate the susceptibility for recurrent AKI or development of chronic kidney disease. In this study, mouse kidneys were subjected to ischemia-reperfusion injury, and the contralateral kidneys remained uninjured to enable comparison and assess injury-induced changes in the kidney proteome. A ZenoTOF 7600 mass spectrometer was optimized for data-independent acquisition (DIA) to achieve comprehensive protein identification and quantification. Short microflow gradients and the generation of a deep kidney-specific spectral library allowed for high-throughput, comprehensive protein quantification. Upon AKI, the kidney proteome was completely remodeled, and over half of the 3945 quantified protein groups changed significantly. Downregulated proteins in the injured kidney were involved in energy production, including numerous peroxisomal matrix proteins that function in fatty acid oxidation, such as ACOX1, CAT, EHHADH, ACOT4, ACOT8, and Scp2. Injured kidneys exhibited severely damaged tissues and injury markers. The comprehensive and sensitive kidney-specific DIA-MS assays feature high-throughput analytical capabilities to achieve deep coverage of the kidney proteome, and will serve as useful tools for developing novel therapeutics to remediate kidney function.
Collapse
Affiliation(s)
- Jordan B Burton
- Buck Institute for Research on Aging, Novato, California, USA
| | - Anne Silva-Barbosa
- Department of Pediatrics, School of Medicine, Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jacob Rose
- Buck Institute for Research on Aging, Novato, California, USA
| | - Katherine Pfister
- Department of Pediatrics, School of Medicine, Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | - Eric S Goetzman
- Department of Pediatrics, School of Medicine, Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sunder Sims-Lucas
- Department of Pediatrics, School of Medicine, Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
17
|
Salzinger B, Lundwall K, Evans M, Mörtberg J, Wallén H, Jernberg T, Kahan T, Lundman P, Tornvall P, Erlinge D, Lindahl B, Baron T, Rezeli M, Spaak J, Jacobson SH. Associations between inflammatory and angiogenic proteomic biomarkers, and cardiovascular events and mortality in relation to kidney function. Clin Kidney J 2024; 17:sfae050. [PMID: 38524235 PMCID: PMC10959071 DOI: 10.1093/ckj/sfae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Indexed: 03/26/2024] Open
Abstract
Background The links between chronic kidney disease (CKD) and the high burden of cardiovascular disease remain unclear. We aimed to explore the association between selected inflammatory and angiogenic biomarkers, kidney function and long-term outcome in patients with an acute coronary syndrome (ACS) and to test the hypothesis that CKD status modifies this association. Methods A total of 1293 ACS patients hospitalized between 2008 and 2015 were followed until 31 December 2017. Plasma was collected on days 1-3 after admission. A total of 13 biomarkers were a priori identified and analysed with two proteomic methods, proximity extension assay or multiple reaction monitoring mass spectrometry. Boxplots and multiple linear regression models were used to study associations between biomarkers and kidney function and adjusted standardized Cox regression with an interaction term for CKD was used to assess whether CKD modified the association between biomarkers and major adverse cardiovascular events and death (MACE+). Results The concentrations of nine biomarkers-endothelial cell-specific molecule-1 (ESM-1), fibroblast growth factor 23 (FGF-23), fractalkine (CX3CL1), interleukin-1 receptor antagonist (IL-1RA), interleukin-18 (IL-18), monocyte chemotactic protein-1 (MCP-1), placenta growth factor (PlGF), transmembrane immunoglobulin 1 (TIM-1) and vascular endothelial growth factor A (VEGFA)-were inversely associated with kidney function. ESM-1, FGF-23 and TIM-1 showed associations with MACE+. Only FGF23 remained independently associated after adjustment for the other biomarkers (hazard ratio per standard deviation increase 1.34; 95% Bonferroni corrected confidence interval 1.19-1.50). None of the biomarkers showed an interaction with CKD. Conclusions The concentrations of 9 of the 13 prespecified inflammatory and angiogenic proteomic biomarkers increased when kidney function declined. Only FGF-23 demonstrated an independent association with MACE+, and this association was not modified by CKD status. These findings further support FGF-23 as an independent prognostic marker in ACS patients with and without CKD.
Collapse
Affiliation(s)
- Barbara Salzinger
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Kristina Lundwall
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Marie Evans
- ME Renal Medicine, Department of Clinical Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Josefin Mörtberg
- Division of Nephrology, Department of Internal Medicine, Centre for Clinical Research, County of Vastmanland and Uppsala University, Uppsala, Sweden
| | - Håkan Wallén
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Tomas Jernberg
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Thomas Kahan
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Pia Lundman
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Per Tornvall
- Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institute, Stockholm, Sweden
| | - David Erlinge
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Bertil Lindahl
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden
| | - Tomasz Baron
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden
| | - Melinda Rezeli
- Clinical Protein Science & Imaging, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Jonas Spaak
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Stefan H Jacobson
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
18
|
Bufkin KB, Karim ZA, Silva J. Review of the limitations of current biomarkers in acute kidney injury clinical practices. SAGE Open Med 2024; 12:20503121241228446. [PMID: 38322582 PMCID: PMC10846001 DOI: 10.1177/20503121241228446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024] Open
Abstract
Acute kidney injury is a prevalent disease in hospitalized patients and is continuously increasing worldwide. Various efforts have been made to define and classify acute kidney injury to understand the progression of this disease. Furthermore, deviations from structure and kidney function and the current diagnostic guidelines are not adequately placed due to baseline serum creatinine values, which are rarely known and estimated based on glomerular function rate, resulting in misclassification of acute kidney injury staging. Hence, the current guidelines are still developing to improve and understand the clinical implications of risk factors and earlier predictive biomarkers of acute kidney injury. Yet, studies have indicated disadvantages and limitations with the current acute kidney injury biomarkers, including lack of sensitivity and specificity. Therefore, the present narrative review brings together the most current evidenced-based practice and literature associated with the limitations of the gold standard for acute kidney injury diagnoses, the need for novel acute kidney injury biomarkers, and the process for biomarkers to be qualified for diagnostic use under the following sections and themes. The introduction section situates the anatomy and normal and abnormal kidney functions related to acute kidney injury disorders. Guidelines in providing acute kidney injury definitions and classification are then considered, followed by a discussion of the disadvantages of standard markers used to diagnose acute kidney injury. Characteristics of an ideal acute kidney injury biomarker are discussed concerning sensitivity, specificity, and anatomic location of injury. A particular focus on the role and function of emerging biomarkers is discussed in relation to their applications and significance to the prognosis and severity of acute kidney injury. Findings show emerging markers are early indicators of acute kidney injury prediction in different clinical settings. Finally, the process required for a biomarker to be applied for diagnostic use is explained.
Collapse
Affiliation(s)
- Kendra B Bufkin
- Department of Interdisciplinary Health Science, College of Allied Health Science, Augusta University, Augusta, GA, USA
| | - Zubair A Karim
- Department of Interdisciplinary Health Science, College of Allied Health Science, Augusta University, Augusta, GA, USA
| | - Jeane Silva
- Department of Health Management, Economics and Policy, Augusta University, Augusta, GA, USA
| |
Collapse
|
19
|
Kiernan EA, Hu D, Philbrook HT, Ix JH, Bonventre JV, Coca SG, Moledina DG, Fried LF, Shlipak MG, Parikh CR. Urinary Biomarkers and Kidney Injury in VA NEPHRON-D: Phenotyping Acute Kidney Injury in Clinical Trials. Am J Kidney Dis 2024; 83:151-161. [PMID: 37726051 PMCID: PMC10841767 DOI: 10.1053/j.ajkd.2023.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 06/23/2023] [Accepted: 07/02/2023] [Indexed: 09/21/2023]
Abstract
RATIONALE & OBJECTIVE Urinary biomarkers of injury, inflammation, and repair may help phenotype acute kidney injury (AKI) observed in clinical trials. We evaluated the differences in biomarkers between participants randomized to monotherapy or to combination renin-angiotensin-aldosterone system (RAAS) blockade in VA NEPHRON-D, where an increased proportion of observed AKI was acknowledged in the combination arm. STUDY DESIGN Longitudinal analysis. SETTING & PARTICIPANTS A substudy of the VA NEPHRON-D trial. PREDICTOR Primary exposure was the treatment arm (combination [RAAS inhibitor] vs monotherapy). AKI is used as a stratifying variable. OUTCOME Urinary biomarkers, including albumin, EGF (epidermal growth factor), MCP-1 (monocyte chemoattractant protein-1), YKL-40 (chitinase 3-like protein 1), and KIM-1 (kidney injury molecule-1). ANALYTICAL APPROACH Biomarkers measured at baseline and at 12 months in trial participants were compared between treatment groups and by AKI. AKI events occurring during hospitalization were predefined safety end points in the original trial. The results were included in a meta-analysis with other large chronic kidney disease trials to assess global trends in biomarker changes. RESULTS In 707 participants followed for a median of 2.2 years, AKI incidence was higher in the combination (20.7%) versus the monotherapy group (12.7%; relative risk [RR], 1.64 [95% CI, 1.16-2.30]). Compared with the monotherapy arm, in the combination arm the urine biomarkers at 12 months were either unchanged (MCP-1: RR, -3% [95% CI, -13% to 9%], Padj=0.8; KIM-1: RR, -10% [95% CI, -20% to 1%], Padj=0.2; EGF, RR-7% [95% CI, -12% to-1%], Padj=0.08) or lower (albuminuria: RR, -24% [95% CI, -37% to-8%], Padj=0.02; YKL: RR, -40% to-44% [95% CI, -58% to-25%], Padj<0.001). Pooled meta-analysis demonstrated reduced albuminuria in the intervention arm across 3 trials and similar trajectories in other biomarkers. LIMITATIONS Biomarker measurement was limited to 2 time points independent of AKI events. CONCLUSIONS Despite the increased risk of serum creatinine-defined AKI, combination RAAS inhibitor therapy was associated with unchanged or decreased urinary biomarkers at 12 months. This suggests a possible role for kidney biomarkers to further characterize kidney injury in clinical trials. PLAIN-LANGUAGE SUMMARY The VA NEPHRON-D trial investigated inhibition of the renin-angiotensin-aldosterone system (RAAS) hormonal axis on kidney outcomes in a large population of diabetic chronic kidney disease patients. The trial was stopped early due to increased events of serum creatinine-defined acute kidney injury in the combination therapy arm. Urine biomarkers can serve as an adjunct to serum creatinine in identifying kidney injury. We found that urinary biomarkers in the combination therapy group were not associated with a pattern of harm and damage to the kidney, despite the increased number of kidney injury events in that group. This suggests that serum creatinine alone may be insufficient for defining kidney injury and supports further exploration of how other biomarkers might improve identification of kidney injury in clinical trials.
Collapse
Affiliation(s)
- Elizabeth A Kiernan
- Division of Nephrology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - David Hu
- Division of Nephrology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Heather Thiessen Philbrook
- Division of Nephrology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Joachim H Ix
- Division of Nephrology-Hypertension, University of California-San Diego, San Diego, California; Veterans Affairs San Diego Healthcare System, San Diego, CA
| | | | - Steven G Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dennis G Moledina
- Section of Nephrology and Clinical and Translational Research Accelerator, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut
| | - Linda F Fried
- Renal Section, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Michael G Shlipak
- Kidney Health Research Collaborative, Department of Medicine, University of California-San Francisco, San Francisco, California
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
20
|
Rossiter A, La A, Koyner JL, Forni LG. New biomarkers in acute kidney injury. Crit Rev Clin Lab Sci 2024; 61:23-44. [PMID: 37668397 DOI: 10.1080/10408363.2023.2242481] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/14/2023] [Accepted: 07/26/2023] [Indexed: 09/06/2023]
Abstract
Acute kidney injury (AKI) is a commonly encountered clinical syndrome. Although it often complicates community acquired illness, it is more common in hospitalized patients, particularly those who are critically ill or who have undergone major surgery. Approximately 20% of hospitalized adult patients develop an AKI during their hospital care, and this rises to nearly 60% in the critically ill, depending on the population being considered. In general, AKI is more common in older adults, in those with preexisting chronic kidney disease and in those with known risk factors for AKI (including diabetes and hypertension). The development of AKI is associated with an increase in both mortality and morbidity, including the development of post-AKI chronic kidney disease. Currently, AKI is defined by a rise in serum creatinine from either a known or derived baseline value and/or oliguria or anuria. However, clinicians may fail to recognize the initial development of AKI because of a delay in the rise of serum creatinine or because of inaccurate urine output monitoring. This, in turn, delays any putative measures to treat AKI or to limit its degree. Consequently, efforts have focused on new biomarkers associated with AKI that may allow early recognition of this syndrome with the intent that this will translate into improved patient outcomes. Here we outline current biomarkers associated with AKI and explore their potential in aiding diagnosis, understanding the pathophysiology and directing therapy.
Collapse
Affiliation(s)
- Adam Rossiter
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
| | - Ashley La
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Jay L Koyner
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Lui G Forni
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
- School of Medicine, Department of Clinical & Experimental Medicine, Faculty of Health Sciences, University of Surrey, Surry, UK
| |
Collapse
|
21
|
Ito S, Yamatani F, Arai Y, Manabe E, Tsujino T. Dimethyl Fumarate Ameliorated Cardiorenal Anemia Syndrome and Improved Overall Survival in Dahl/Salt-Sensitive Rats. J Pharmacol Exp Ther 2023; 387:299-305. [PMID: 37857438 DOI: 10.1124/jpet.123.001692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 10/21/2023] Open
Abstract
Cardiovascular disease, chronic kidney disease, and anemia are known to adversely affect each other. Inflammation is commonly involved in these diseases. Cardiorenal anemia syndrome (CRAS) is the name given to this mutually harmful condition. Dimethyl fumarate (DMF) is a Food and Drug Administration-approved antioxidant and anti-inflammatory agent. The purpose of this study was to investigate the effects of DMF on Dahl/salt-sensitive (DS) rats as a CRAS model. Six-week-old DS rats were divided into three groups: the control group, the high-salt (HS) group, and the HS+DMF group. The HS and HS+DMF groups were fed a high-salt diet (8% NaCl) from 6 weeks of age. In the HS+DMF group, DMF (90 mg/kg per day) was orally administered from 6 to 15 weeks of age. Systolic blood pressure was measured every 2 weeks. The heart and renal injuries were assessed with histopathological analysis. The heart and renal expression of mRNAs was assessed by reverse-transcription polymerase chain reaction. DMF significantly improved overall survival, which was shortened by HS in DS rats. Systolic blood pressure increased in the HS group compared with the control group, and DMF tended to suppress this change. DMF ameliorated the cardiac and renal abnormalities confirmed in the HS group by histopathological analysis. Furthermore, the changes in mRNA expressions associated with disease exacerbation in the HS group were suppressed by DMF. DMF also improved anemia. This study suggests that DMF improves overall survival in DS rats through organ-protective effects and is effective against cardiorenal anemia syndrome. SIGNIFICANCE STATEMENT: Dimethyl fumarate was found to improve overall survival in Dahl/salt-sensitive rats, associated with its ability to ameliorate anemia and induce cardioprotective and renoprotective effects through anti-inflammatory and antifibrotic effects.
Collapse
Affiliation(s)
- Satoyasu Ito
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Fuyuka Yamatani
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Yuri Arai
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Eri Manabe
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Takeshi Tsujino
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| |
Collapse
|
22
|
Elbarbary NS, Ismail EAR, Mohamed SA. Omega-3 fatty acids supplementation improves early-stage diabetic nephropathy and subclinical atherosclerosis in pediatric patients with type 1 diabetes: A randomized controlled trial. Clin Nutr 2023; 42:2372-2380. [PMID: 37862823 DOI: 10.1016/j.clnu.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/21/2023] [Accepted: 10/07/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Numerous studies have evaluated the beneficial effects of omega-3 fatty acids on inflammatory, autoimmune and renal diseases. However, data about the effects of omega-3 fatty acids on diabetic kidney disease in type 1 diabetes mellitus (T1DM) are lacking. OBJECTIVES This randomized-controlled trial assessed the effect of oral omega-3 supplementation on glycemic control, lipid profile, albuminuria level, kidney injury molecule-1 (KIM-1) and carotid intima media thickness (CIMT) in pediatric patients with T1DM and diabetic nephropathy. METHODS Seventy T1DM patients and diabetic nephropathy were enrolled with a mean age 15.2 ± 1.96 years and median disease duration 7 years. Patients were randomly assigned into two groups; intervention group which received oral omega-3 fatty acids capsules (1 g daily). The other group received a matching placebo and served as a control group. Both groups were followed-up for 6 months with assessment of fasting blood glucose (FBG), HbA1c, fasting lipids, urinary albumin creatinine ratio (UACR), KIM-1 and CIMT. RESULTS After 6 months, omega-3 fatty acids adjuvant therapy for the intervention group resulted in a significant decrease in FBG, HbA1c, triglycerides, total cholesterol, LDL-cholesterol, UACR, KIM-1 and CIMT, whereas, HDL-cholesterol was significantly higher post-therapy compared with baseline levels and compared with the control group (p < 0.05). Baseline KIM-1 levels were positively correlated to HbA1c, UACR and CIMT. Supplementation with omega-3 fatty acids was safe and well-tolerated. CONCLUSIONS Omega-3 fatty acids as an adjuvant therapy in pediatric T1DM patients with diabetic nephropathy improved glycemic control, dyslipidemia and delayed disease progression and subclinical atherosclerosis among those patients. This trial was registered under ClinicalTrials.gov Identifier no. NCT05980026.
Collapse
|
23
|
Fadallah M, Abdelhalim A, Hashem A, Mortada WI, Ibrahim HAM, Sheir KZ, Harraz AM, El-Kenawy MR, El-Nahas AR. The Ideal Interval Between Repeated Shockwaves Lithotripsy Sessions for Renal Stones: A Randomized Controlled Trial. J Endourol 2023; 37:1305-1313. [PMID: 37767632 DOI: 10.1089/end.2023.0345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
Objectives: To assess the ideal interval between repeated extracorporeal shockwave lithotripsy (SWL) for renal stones. Patient and Methods: Eligible patients with a single renal stone ≤20 mm who required SWL were randomly assigned to one of three groups based on intervals between first and second sessions. Patients underwent the second session after 3, 7, and 14 days in Groups 1, 2, and 3, respectively. Tubular functions were assessed through comparisons of urinary execration of kidney injury molecule-1 (KIM-1), neutrophil gelatinase associated lipocalin (NGAL), and interleukin-18 (IL-18) with pre-SWL values, whereas glomerular function was assessed by comparisons of protein/creatinine ratio with pre-SWL and changes in ipsilateral renal function on isotope scans. Treatment success was assessed by noncontrast CT after 3 months. Results: All demographics of the 166 patients included in the study were comparable between the three groups. There were significant elevations of tubular biomarkers and protein/creatinine ratio after first and second SWL sessions compared with pre-SWL values (p < 0.0001). All tubular biomarkers returned to pre-SWL values at 7 and 14 days after second session, whereas they remained significantly elevated 3 days after second session (p = 0.027, < 0.001 and <0.001 for KIM-1, NGAL, and IL-18, respectively). SWL success was 73.6% in Group 1, 83.7% in Group 2, and 81% in Group 3. A significant decrease in ipsilateral renal split function was observed in Group 1 at the 3-month follow-up. Conclusions: An interval of 7 days is required between SWL sessions when treating renal stones to allow for complete recovery of kidney functions. Clinical Trial Registration: ID: NCT04575480.
Collapse
Affiliation(s)
- Mohamed Fadallah
- Urology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ahmed Abdelhalim
- Urology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
- Department of Urology, West Virginia University, Morgantown, West Virginia, USA
| | - Abdelwahab Hashem
- Urology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Wael I Mortada
- Laboratory Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Hadeer A M Ibrahim
- Laboratory Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Khaled Z Sheir
- Urology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ahmed M Harraz
- Urology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Mahmoud R El-Kenawy
- Urology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ahmed R El-Nahas
- Urology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
24
|
Fan F, Xu P. Global biomarkers trends in acute kidney injury: a bibliometric analysis. Ren Fail 2023; 45:2278300. [PMID: 37994407 PMCID: PMC11001340 DOI: 10.1080/0886022x.2023.2278300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/28/2023] [Indexed: 11/24/2023] Open
Abstract
OBJECTIVES Acute kidney injury (AKI) is a common global condition with high morbidity and mortality rates. Biomarkers can aid in the diagnosis, prediction, intervention, and outcome assessment of AKI. This study aimed to summarize the current research status and identify research hotspots for AKI biomarkers using bibliometric analysis. METHODS Relevant original English language articles were retrieved from the Science Citation Index Expanded of the Web of Science Core Collection database, from inception to 31 December 2022. Full records and related cited references from all the documents were collected and analyzed. RESULTS A total of 16368 authors from 3379 institutions in 83 countries/regions contributed to 2916 documents that were published in 712 academic journals. Annual publication output followed exponential growth since 2008. The United States, the University of Pittsburgh, and the American Journal of Physiology-Renal Physiology were the most productive countries, institutions, and journals in terms of research outputs, respectively. The area of interest has shifted from neutrophil gelatinase-associated lipocalin, cell cycle, and tubular damage toward sepsis and COVID-19. Apoptosis, inflammation, and chronic kidney disease have become popular in recent years, and studies on ferroptosis, machine learning, COVID-19, and renal fibrosis will be the focus of future research. IMPLICATIONS This bibliometric study suggests that future research on AKI biomarkers would focus on ferroptosis, renal fibrosis and COVID-19. Artificial intelligence, such as machine learning, maybe the most promising direction for the discovery and validation of AKI biomarkers.
Collapse
Affiliation(s)
- Fan Fan
- Department of General Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Peifeng Xu
- Department of General Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
25
|
Campbell RE, Chen CH, Edelstein CL. Overview of Antibiotic-Induced Nephrotoxicity. Kidney Int Rep 2023; 8:2211-2225. [PMID: 38025228 PMCID: PMC10658282 DOI: 10.1016/j.ekir.2023.08.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 12/01/2023] Open
Abstract
Drug-induced nephrotoxicity accounts for up to 60% of cases of acute kidney injury (AKI) in hospitalized patients and is associated with increased morbidity and mortality in both adults and children. Antibiotics are one of the most common causes of drug-induced nephrotoxicity. Mechanisms of antibiotic-induced nephrotoxicity include glomerular injury, tubular injury or dysfunction, distal tubular obstruction from casts, and acute interstitial nephritis (AIN) mediated by a type IV (delayed-type) hypersensitivity response. Clinical manifestations of antibiotic-induced nephrotoxicity include acute tubular necrosis (ATN), AIN, and Fanconi syndrome. Given the potential nephrotoxic effects of antibiotics on critically ill patients, the use of novel biomarkers can provide information to optimize dosing and duration of treatment and can help prevent nephrotoxicity when traditional markers, such as creatinine, are unreliable. Use of novel kidney specific biomarkers, such as cystatin C and urinary kidney injury molecule-1 (KIM-1), may result in earlier detection of AKI, dose adjustment, or discontinuation of antibiotic and development of nonnephrotoxic antibiotics.
Collapse
Affiliation(s)
- Ruth E. Campbell
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chang Huei Chen
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Charles L. Edelstein
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
26
|
Fatima N, Ali R, Faisal T, Kulkarni K, Patel S, Hussain T. Macrophage angiotensin AT 2 receptor activation is protective against early phases of LPS-induced acute kidney injury. Am J Physiol Renal Physiol 2023; 325:F552-F563. [PMID: 37615049 PMCID: PMC10878726 DOI: 10.1152/ajprenal.00177.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/27/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Lipopolysaccharide (LPS)-induced acute kidney injury (AKI) is characterized by inflammation and infiltration of immune cells, mainly neutrophils and macrophages, and results in sudden renal dysfunction. Previously, we have reported the anti-inflammatory and renoprotective role of the angiotensin II type 2 receptor (AT2R), expressed on kidney tubular cells and immune cells, in LPS-induced AKI. Moreover, in vitro studies revealed macrophage AT2R activation shifts the cells to the anti-inflammatory M2 subtype. However, the protective role of the macrophage AT2R in a model of AKI is unknown. The present study addressed this question by adoptive transfer of bone marrow-derived macrophages (BMDMs) in systemic macrophage-depleted mice. We acquired significant systemic macrophage depletion by two doses of liposomal clodronate (CLD), and the mice were repopulated with BMDMs (CD11b+F4/80+, double positive) primed with AT2R agonist C21 (CLD + MacC21 + LPS) or vehicle (CLD + Mac + LPS) in vitro for 60 min, followed by LPS (5 mg/kg body wt ip) challenge. We observed a gradual increase in the CD11b+ cells at 2 and 24 h after the LPS challenge. However, kidney CD11b+ cells in the CLD + Mac + LPS group were elevated compared with the CLD + MacC21 + LPS group at 2 h after the LPS challenge. The level of inflammatory cytokine (tumor necrosis factor-α) was elevated at 2 h, which was reduced significantly in CLD + MacC21 + LPS-treated animals. Also, CLD + MacC21 + LPS-treated animals had elevated plasma and renal IL-10, indicating an anti-inflammatory role of C21-treated BMDMs. Renal functional injury in CLD + MacC21 + LPS-treated animals was partially improved. Collectively, the data demonstrate that BMDM AT2R stimulation results in anti-inflammation and partial renoprotection against early stages of LPS-induced AKI.NEW & NOTEWORTHY Endotoxin such as lipopolysaccharide (LPS) induces acute kidney injury (AKI), which is a risk factor for and often leads to chronic kidney diseases. The present study revealed that bone marrow-derived macrophage activation of the angiotensin II type 2 receptor (AT2R) contributes to the anti-inflammation and partial renoprotection against early stages of LPS-induced AKI. Since AT2R is an emerging anti-inflammatory and organ-protective target, this study advances our understanding of AT2R's anti-inflammatory mechanisms associated with renoprotection.
Collapse
Affiliation(s)
- Naureen Fatima
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Riyasat Ali
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Tahmid Faisal
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Kalyani Kulkarni
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Sanket Patel
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Tahir Hussain
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| |
Collapse
|
27
|
Ye Q, Xu G, Xue C, Pang S, Xie B, Huang G, Li H, Chen X, Yang R, Li W. Urinary SPP1 has potential as a non-invasive diagnostic marker for focal segmental glomerulosclerosis. FEBS Open Bio 2023; 13:2061-2080. [PMID: 37696527 PMCID: PMC10626280 DOI: 10.1002/2211-5463.13704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/26/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a type of chronic glomerular nephropathy showing characteristic glomerular sclerosis, diagnosed by kidney biopsy. However, it is difficult and expensive to monitor disease progression with repeated renal biopsy in clinical practice, and thus here we explored the feasibility of urine biomarkers as non-invasive diagnostic tools. We downloaded scRNA-seq datasets of 20 urine cell samples and 3 kidney tissues and obtained two gene lists encoding extracellular proteins for bioinformatic analysis; in addition, we identified key EP-Genes by immunohistochemical staining and performed bulk RNA sequencing with 12 urine samples. We report that urine cells and kidney cells were correlated. A total of 64 EP-Genes were acquired by intersecting genes of distal tubular cluster with extracellular proteins. Function enrichment analysis showed that EP-Genes might be involved in the immune response and extracellular components. Six key EP-Genes were identified and correlated with renal function. IMC showed that key EP-Genes were located mainly in tubules. Cross verification and examination of a urine RNAseq dataset showed that SPP1 had diagnostic potential for FSGS. The presence of urine SPP1 was primarily associated with macrophage infiltration in kidney, and the pathogenesis of FSGS may be related to innate immunity. Urinary cells seemed to be strongly similar to kidney cells. In summary, SPP1 levels reflect renal function and may have potential as a biomarker for non-invasive diagnosis of FSGS.
Collapse
Affiliation(s)
- Qinglin Ye
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Guiling Xu
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Chao Xue
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Shuting Pang
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Boji Xie
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Guanwen Huang
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Haoyu Li
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Xuesong Chen
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Rirong Yang
- Centre for Genomic and Personalized MedicineDepartment of ImmunologySchool of Basic Medical SciencesGuangxi Medical UniversityNanning530021China
| | - Wei Li
- Department of NephrologyThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
28
|
Möckel T, Boegel S, Schwarting A. Transcriptome analysis of renal ischemia/reperfusion (I/R) injury in BAFF and BAFF-R deficient mice. PLoS One 2023; 18:e0291619. [PMID: 37751458 PMCID: PMC10522044 DOI: 10.1371/journal.pone.0291619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 09/02/2023] [Indexed: 09/28/2023] Open
Abstract
Acute kidney injury (AKI) accompanies with high morbidity and mortality. Incomplete renal recovery can lead to chronic and finally end-stage kidney disease, which results in the requirement of lifelong dialysis or kidney transplantation. Consequently, finding predictive biomarker and therefore developing preventive therapeutic approaches is an urgent need. For this purpose, a better understanding of the mechanism underlying AKI is necessary. The cytokine BAFF (B cell activating factor) is related to AKI by supporting B cells, which in turn play an important role in inflammatory processes and the production of antibodies. In our study, we investigated the role of BAFF and its receptor BAFF-R in the early phase of AKI. Therefore, we performed the well-established ischemia/reperfusion (I/R) model in BAFF (B6.129S2-Tnfsf13btm1Msc/J) and BAFF-R (B6(Cg)-Tnfrsf13ctm1Mass/J) deficient mice. Transcriptome of ischemic and contralateral control kidneys was analyzed and compared to wildtype littermates. We detected the upregulation of Lcn2, Lyz2, Cd44, Fn1 and Il1rn in ischemic kidneys as well as the downregulation of Kl. Furthermore, we revealed different expression patterns in BAFF and BAFF-R knockout mice. Compared to wildtype littermates, up- and downregulation of each investigated gene were higher in BAFF-R knockout and lower in BAFF knockout. Our findings indicate a positive impact of BAFF knockout in early phase of AKI, while BAFF-R knockout seems to worsen I/R injury. In addition, our study shows for the first time a remarkable renal upregulation of Lyz2 in a murine I/R model. Therefore, we consider Lyz2 as conceivable predictive or early biomarker in case of I/R and AKI.
Collapse
Affiliation(s)
- Tamara Möckel
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sebastian Boegel
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Schwarting
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Center for Rheumatic Disease Rhineland-Palatinate GmbH, Bad Kreuznach, Germany
| |
Collapse
|
29
|
Loke RYJ, Chin CF, Liang G, Wong BH, Galam DLA, Tan BC, Chua GL, Minegishi S, Morisawa N, Sidorov I, Heijs B, Titze J, Wenk MR, Torta F, Silver DL. Mfsd2a-mediated lysolipid transport is important for renal recovery after acute kidney injury. J Lipid Res 2023; 64:100416. [PMID: 37467896 PMCID: PMC10424216 DOI: 10.1016/j.jlr.2023.100416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Acute kidney injury (AKI) is a global public health concern with high mortality and morbidity. In ischemic-reperfusion injury (IRI), a main cause of AKI, the brush border membrane of S3 proximal tubules (PT) is lost to the tubular lumen. How injured tubules reconstitute lost membrane lipids during renal recovery is not known. Here, we identified Mfsd2a, a sodium-dependent lysophosphatidylcholine (LPC) transporter, to be expressed specifically in the basolateral membrane of S3 PT. Using an in vivo activity probe for Mfsd2a, transport activity was found to be specific to the S3 PT. Mice with haploinsufficiency of Mfsd2a exhibited delayed recovery of renal function after acute IRI, with depressed urine osmolality and elevated levels of histological markers of damage, fibrosis, and inflammation, findings corroborated by transcriptomic analysis. Lipidomics revealed a deficiency in docosahexaenoic acid (DHA) containing phospholipids in Mfsd2a haploinsufficiency. Treatment of Mfsd2a haploinsufficient mice with LPC-DHA improved renal function and reduced markers of injury, fibrosis, and inflammation. Additionally, LPC-DHA treatment restored S3 brush border membrane architecture and normalized DHA-containing phospholipid content. These findings indicate that Mfsd2a-mediated transport of LPC-DHA is limiting for renal recovery after AKI and suggest that LPC-DHA could be a promising dietary supplement for improving recovery following AKI.
Collapse
Affiliation(s)
- Randy Y J Loke
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Cheen Fei Chin
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Gao Liang
- Singapore Lipidomics Incubator, Life Sciences Institute, NUS, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Bernice H Wong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Dwight L A Galam
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Bryan C Tan
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Geok-Lin Chua
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Shintaro Minegishi
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Norihiko Morisawa
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Iulia Sidorov
- Center of Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands
| | - Bram Heijs
- Center of Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands
| | - Jens Titze
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, NUS, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, NUS, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, NUS, Singapore
| | - David L Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore.
| |
Collapse
|
30
|
Rasking L, Koshy P, Bongaerts E, Bové H, Ameloot M, Plusquin M, De Vusser K, Nawrot TS. Ambient black carbon reaches the kidneys. ENVIRONMENT INTERNATIONAL 2023; 177:107997. [PMID: 37269720 DOI: 10.1016/j.envint.2023.107997] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND Ultrafine particles, including black carbon (BC), can reach the systemic circulation and therefore may distribute to distant organs upon inhalation. The kidneys may be particularly vulnerable to the adverse effects of BC exposure due to their filtration function. OBJECTIVES We hypothesized that BC particles reach the kidneys via the systemic circulation, where the particles may reside in structural components of kidney tissue and impair kidney function. METHODS In kidney biopsies from 25 transplant patients, we visualized BC particles using white light generation under femtosecond-pulsed illumination. The presence of urinary kidney injury molecule-1 (KIM-1) and cystatin c (CysC) were evaluated with ELISA. We assessed the association between internal and external exposure matrices and urinary biomarkers using Pearson correlation and linear regression models. RESULTS BC particles could be identified in all biopsy samples with a geometric mean (5th, 95th percentile) of 1.80 × 103 (3.65 × 102, 7.50 × 103) particles/mm3 kidney tissue, predominantly observed in the interstitium (100 %) and tubules (80 %), followed by the blood vessels and capillaries (40 %), and the glomerulus (24 %). Independent from covariates and potential confounders, we found that each 10 % higher tissue BC load resulted in 8.24 % (p = 0.03) higher urinary KIM-1. In addition, residential proximity to a major road was inversely associated with urinary CysC (+10 % distance: -4.68 %; p = 0.01) and KIM-1 (+10 % distance: -3.99 %; p < 0.01). Other urinary biomarkers, e.g., the estimated glomerular filtration rate or creatinine clearance showed no significant associations. DISCUSSION AND CONCLUSION Our findings that BC particles accumulate near different structural components of the kidney represent a potential mechanism explaining the detrimental effects of particle air pollution exposure on kidney function. Furthermore, urinary KIM-1 and CysC show potential as air pollution-induced kidney injury biomarkers for taking a first step in addressing the adverse effects BC might exert on kidney function.
Collapse
Affiliation(s)
- Leen Rasking
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Priyanka Koshy
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Eva Bongaerts
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Hannelore Bové
- Department of Sciences, Hasselt University, Diepenbeek, Belgium
| | - Marcel Ameloot
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Katrien De Vusser
- Nephrology and Kidney Transplantation, University Hospital Leuven, Leuven, Belgium; Department of Microbiology and Immunology, Leuven University, Leuven, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium; Department of Public Health and Primary Care, Environment and Health Unit, Leuven University, Leuven, Belgium.
| |
Collapse
|
31
|
Ma J, Wang X, Gu R, Guo D, Shi C, Kollisch-Singule M, Suo L, Luo J, Meng Q, Cooney RN. PROPHYLACTIC n CMT-3 ATTENUATES SEPSIS-INDUCED ACUTE KIDNEY INJURY IN ASSOCIATION WITH NLRP3 INFLAMMASOME ACTIVATION AND APOPTOSIS. Shock 2023; 59:922-929. [PMID: 36939682 PMCID: PMC10205665 DOI: 10.1097/shk.0000000000002118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
ABSTRACT Background: The kidney is the most common extrapulmonary organ injured in sepsis. The current study examines the ability of aerosolized nanochemically modified tetracycline 3 (nCMT-3), a pleiotropic anti-inflammatory agent, to attenuate acute kidney injury (AKI) caused by intratracheal LPS. Methods: C57BL/6 mice received aerosolized intratracheal nCMT-3 (1 mg/kg) or saline, followed by intratracheal LPS (2.5 mg/kg) to induce acute lung injury-induced AKI. Tissues were harvested at 24 h. The effects of nCMT-3 and LPS on AKI were assessed by plasma/tissue levels of serum urea nitrogen, creatinine, neutrophil gelatinase-associated lipocalin, kidney injury molecule 1, and renal histology. Renal matrix metalloproteinase (MMP) level/activity, cytochrome C, Bax, Bcl-2, caspase-3, p38 mitogen-activated protein kinase activation, NLRP3, and caspase-1 were also measured. Apoptotic cells in kidney were determined by TUNEL assay. Renal levels of IL-1β and IL-6 were measured to assess inflammation. Results: Acute lung injury-induced AKI was characterized by increased plasma blood urea nitrogen, creatinine, injury biomarkers (neutrophil gelatinase-associated lipocalin, kidney injury molecule 1), and histologic evidence of renal injury. Lipopolysaccharide-treated mice demonstrated renal injury with increased levels of inflammatory cytokines (IL-1β, IL-6), active MMP-2 and MMP-9, proapoptotic proteins (cytochrome C, Bax/Bcl-2 ratio, cleaved caspase-3), apoptotic cells, inflammasome activation (NLRP3, caspase-1), and p38 signaling. Intratracheal nCMT-3 significantly attenuated all the measured markers of renal injury, inflammation, and apoptosis. Conclusions: Pretreatment with aerosolized nCMT-3 attenuates LPS-induced AKI by inhibiting renal NLRP3 inflammasome activation, renal inflammation, and apoptosis.
Collapse
Affiliation(s)
- Julia Ma
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Xiaojing Wang
- Department Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Raymond Gu
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Dandan Guo
- Department Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Changying Shi
- Department Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Michaela Kollisch-Singule
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
- Department Sepsis Interdisciplinary Research Center (SIRC), State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Liye Suo
- Department Pathology, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Juntao Luo
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
- Department Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
- Department Sepsis Interdisciplinary Research Center (SIRC), State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Qinghe Meng
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
- Department Sepsis Interdisciplinary Research Center (SIRC), State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| | - Robert N Cooney
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
- Department Sepsis Interdisciplinary Research Center (SIRC), State University of New York (SUNY), Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
32
|
Joshi MD, Iacoban P, Scheetz MH. Pharmacokinetic and Biomarker Quantification Studies on Vancomycin-Loaded PEGylated Liposomes and Its Potential to Reduce Vancomycin-Induced Kidney Injury: A Rat Study. Pharmaceutics 2023; 15:1582. [PMID: 37376031 DOI: 10.3390/pharmaceutics15061582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Vancomycin is a commonly used antibiotic in hospital settings, especially against Methicillin-resistant staphylococcus aureus (MRSA). One of the major adverse events of vancomycin use in adults is kidney injury. The drug concentration, specifically the area under the concentration curve, predicts kidney injury in adults receiving vancomycin. To attempt to reduce vancomycin-induced nephrotoxicity, we have successfully encapsulated vancomycin in polyethylene glycol-coated liposomes (PEG-VANCO-lipo). We have previously carried out in vitro cytotoxicity studies on kidney cells using PEG-VANCO-lipo and found it to be minimally toxic compared to the standard vancomycin. In this study, we have dosed male adult rats with PEG-VANCO-lipo or vancomycin HCl and compared plasma vancomycin concentrations and KIM-1 as an injury biomarker in rat urine. Male Sprague Dawley rats (350 ± 10 g) were administered vancomycin (n = 6) or PEG-VANCO-lipo (n = 6) 150 mg/kg/day for three days using an IV infusion in the left jugular vein catheter. Blood was collected for plasma at 15, 30, 60, 120, 240, and 1440 min after the first and the last IV dose. Urine was collected 0-2, 2-4, 4-8, and 8-24 h after the first and the last IV infusions using metabolic cages. The animals were observed for three days after the last compound administration. Vancomycin was quantified in plasma by LC-MS/MS. Urinary KIM-1 analysis was done by using an ELISA kit. Three days after the last dose, under terminal anesthesia with IP ketamine (65-100 mg/kg) and xylazine (7-10 mg/kg), rats were euthanized. Vancomycin urine and kidney concentrations and KIM-1 were lower on day three in the PEG-Vanco-lipo group compared to the vancomycin group (p < 0.05, ANOVA and/or t-test). There was a significant reduction in plasma vancomycin concentration on day one and day three (p < 0.05, t-test) in the vancomycin group compared to the PEG-VANCO-lipo group. Vancomycin-loaded PEGylated liposomes resulted in lower levels of kidney injury, as noted by a decrease in KIM-1 values. Moreover, longer circulation in plasma with increased concentration in plasma as opposed to the kidney was observed with the PEG-VANCO-lipo group. The results indicate the high potential of PEG-VANCO-lipo in decreasing the nephrotoxicity of vancomycin clinically.
Collapse
Affiliation(s)
- Medha D Joshi
- College of Pharmacy, Midwestern University, Glendale Campus, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Paulina Iacoban
- College of Pharmacy, Midwestern University, Glendale Campus, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Marc H Scheetz
- College of Pharmacy, Midwestern University, Downers Grove Campus, 555 31st Street, Downers Grove, IL 60515, USA
| |
Collapse
|
33
|
Shati AA, Alkabli J, Alfaifi MY, Elbehairi SEI, Elshaarawy RFM, Serag WM, Hassan YA. Comparison of the ameliorative roles of crab chitosan nanoparticles and mesenchymal stem cells against cisplatin-triggered nephrotoxicity. Int J Biol Macromol 2023:124985. [PMID: 37230447 DOI: 10.1016/j.ijbiomac.2023.124985] [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: 12/15/2022] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
AIM In the present investigation, we compared the effects of mesenchymal stem cells extracted from bone marrow (BMSCs) and crab chitosan nanoparticles (CCNPs) on renal fibrosis in cisplatin (CDDP)-induced kidney injury rats. MATERIAL AND METHODS 90 male Sprague-Dawley (SD) rats were divided into two equal groups and alienated. Group I was set into three subgroups: the control subgroup, the CDDP-infected subgroup (acute kidney injury), and the CCNPs-treated subgroup. Group II was also divided into three subgroups: the control subgroup, the CDDP-infected subgroup (chronic kidney disease), and the BMSCs-treated subgroup. Through biochemical analysis and immunohistochemical research, the protective effects of CCNPs and BMSCs on renal function have been identified. RESULTS CCNPs and BMSC treatment resulted in a substantial rise in GSH and albumin and a decrease in KIM-1, MDA, creatinine, urea, and caspase-3 when compared to the infected groups (p < 0.05). CONCLUSION According to the current research, chitosan nanoparticles and BMSCs may be able to reduce renal fibrosis in acute and chronic kidney diseases caused by CDDP administration, with more improvement of kidney damage resembling normal cells after CCNPs administration.
Collapse
Affiliation(s)
- Ali A Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - J Alkabli
- Department of Chemistry, College of Sciences and Arts - Alkamil, University of Jeddah, Jeddah 23218, Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Serag Eldin I Elbehairi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia; Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El-Zeraa St., Agouza, Giza, Egypt
| | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt; Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany.
| | - Waleed M Serag
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt
| | - Yasser A Hassan
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
34
|
Diao HY, Zhu W, Liu J, Yin S, Wang JH, Li CL. Salvianolic Acid A Improves Rat Kidney Injury by Regulating MAPKs and TGF-β1/Smads Signaling Pathways. Molecules 2023; 28:3630. [PMID: 37110864 PMCID: PMC10144349 DOI: 10.3390/molecules28083630] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Salvianolic acid A (SAA) is one of the major components in Salvia miltiorrhiza Bge., with various pharmacological activities, and is likely to be a promising agent for the treatment of kidney diseases. The purpose of this study was to explore the protective effect and mechanisms of SAA on kidney disease. In this study, the improvement effects of SAA (10, 20, 40 mg/kg, i.g.) on kidney injury rats were investigated by detecting the levels of KIM-1, NGAL in serum and UP in the urine of AKI model rats established with gentamicin, as well as the levels of SCr and UREA in serum and IL-6, IL-12, MDA and T-SOD in the kidneys of CKD model rats established with 5/6 nephrectomy. HE and Masson staining were used to observe the histopathological changes in the kidney. Network pharmacology and Western blotting were used to explore the mechanism of SAA in improving kidney injury. The results showed that SAA improved kidney function in kidney injury rats by reducing the kidney index and pathological injury by HE and Masson staining, reducing the levels of KIM-1, NGAL and UP in AKI rats and UREA, SCr and UP in CKD rats, as well as exerting anti-inflammatory and anti-oxidative stress effects by inhibiting the release of IL-6 and IL-12, reducing MDA and increasing T-SOD. Western blotting results showed that SAA significantly reduced the phosphorylation levels of ERK1/2, p38, JNK and smad2/3, and the expression of TLR-4 and smad7. In conclusion, SAA plays a significant role in improving kidney injury in rats and the mechanism may be achieved by regulating the MAPKs and TGF-β1/smads signaling pathways.
Collapse
Affiliation(s)
- Hai-Yang Diao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wei Zhu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jie Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Sheng Yin
- Department of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin-Hui Wang
- Department of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chun-Li Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
35
|
Li N, Lin G, Zhang H, Sun J, Gui M, Liu Y, Li W, Zhan Z, Li Y, Pan S, Liu J, Tang J. Lyn attenuates sepsis-associated acute kidney injury by inhibition of phospho-STAT3 and apoptosis. Biochem Pharmacol 2023; 211:115523. [PMID: 37003346 DOI: 10.1016/j.bcp.2023.115523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a life-threatening condition associated with high mortality and morbidity. However, the underlying pathogenesis of SA-AKI is still unclear. Lyn belongs to Src family kinases (SFKs), which exert numerous biological functions including modulation in receptor-mediated intracellular signaling and intercellular communication. Previous studies demonstrated that Lyn gene deletion obviously aggravates LPS-induced lung inflammation, but the role and possible mechanism of Lyn in SA-AKI have not been reported yet. Here, we found that Lyn protected against renal tubular injury in cecal ligation and puncture (CLP) induced AKI mouse model by inhibition of signal transducer and activator of transcription 3 (STAT3) phosphorylation and cell apoptosis. Moreover, Lyn agonist MLR-1023 pretreatment improved renal function, inhibited STAT3 phosphorylation and decreased cell apoptosis. Thus, Lyn appears to play a crucial role in orchestrating STAT3-mediated inflammation and cell apoptosis in SA-AKI. Hence, Lyn kinase may be a promising therapeutic target for SA-AKI.
Collapse
Affiliation(s)
- Nannan Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Guoxin Lin
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Jian Sun
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Ming Gui
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Yan Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Wei Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Zishun Zhan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Yisu Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Shiqi Pan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Jishi Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Juan Tang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
36
|
Huang J, Caliskan Guzelce E, Gholami SK, Gawelek KL, Mitchell RN, Pojoga LH, Romero JR, Williams GH, Adler GK. Effects of Mineralocorticoid Receptor Blockade and Statins on Kidney Injury Marker 1 (KIM-1) in Female Rats Receiving L-NAME and Angiotensin II. Int J Mol Sci 2023; 24:6500. [PMID: 37047470 PMCID: PMC10095483 DOI: 10.3390/ijms24076500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
Kidney injury molecule-1 (KIM-1) is a biomarker of renal injury and a predictor of cardiovascular disease. Aldosterone, via activation of the mineralocorticoid receptor, is linked to cardiac and renal injury. However, the impact of mineralocorticoid receptor activation and blockade on KIM-1 is uncertain. We investigated whether renal KIM-1 is increased in a cardiorenal injury model induced by L-NAME/ANG II, and whether mineralocorticoid receptor blockade prevents the increase in KIM-1. Since statin use is associated with lower aldosterone, we also investigated whether administering eiSther a lipophilic statin (simvastatin) or a hydrophilic statin (pravastatin) prevents the increase in renal KIM-1. Female Wistar rats (8-10 week old), consuming a high salt diet (1.6% Na+), were randomized to the following conditions for 14 days: control; L-NAME (0.2 mg/mL in drinking water)/ANG II (225 ug/kg/day on days 12-14); L-NAME/ANG II + eplerenone (100 mg/kg/day p.o.); L-NAME/ANG II + pravastatin (20 mg/kg/day p.o.); L-NAME/ANG II + simvastatin (20 mg/kg/day p.o.). Groups treated with L-NAME/ANG II had significantly higher blood pressure, plasma and urine aldosterone, cardiac injury/stroke composite score, and renal KIM-1 than the control group. Both eplerenone and simvastatin reduced 24-h urinary KIM-1 (p = 0.0046, p = 0.031, respectively) and renal KIM-1 immunostaining (p = 0.004, p = 0.037, respectively). Eplerenone also reduced renal KIM-1 mRNA expression (p = 0.012) and cardiac injury/stroke composite score (p = 0.04). Pravastatin did not affect these damage markers. The 24-h urinary KIM-1, renal KIM-1 immunostaining, and renal KIM-1 mRNA expression correlated with cardiac injury/stroke composite score (p < 0.0001, Spearman ranked correlation = 0.69, 0.66, 0.59, respectively). In conclusion, L-NAME/ANG II increases renal KIM-1 and both eplerenone and simvastatin blunt this increase in renal KIM-1.
Collapse
Affiliation(s)
- Jiayan Huang
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ezgi Caliskan Guzelce
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shadi K. Gholami
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kara L. Gawelek
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Richard N. Mitchell
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Luminita H. Pojoga
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jose R. Romero
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gordon H. Williams
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gail K. Adler
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
37
|
Burton JB, Silva-Barbosa A, Bons J, Rose J, Pfister K, Simona F, Gandhi T, Reiter L, Bernhardt O, Hunter CL, Goetzman ES, Sims-Lucas S, Schilling B. Substantial Downregulation of Mitochondrial and Peroxisomal Proteins during Acute Kidney Injury revealed by Data-Independent Acquisition Proteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.26.530107. [PMID: 36865241 PMCID: PMC9980295 DOI: 10.1101/2023.02.26.530107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Acute kidney injury (AKI) manifests as a major health concern, particularly for the elderly. Understanding AKI-related proteome changes is critical for prevention and development of novel therapeutics to recover kidney function and to mitigate the susceptibility for recurrent AKI or development of chronic kidney disease. In this study, mouse kidneys were subjected to ischemia-reperfusion injury, and the contralateral kidneys remained uninjured to enable comparison and assess injury-induced changes in the kidney proteome. A fast-acquisition rate ZenoTOF 7600 mass spectrometer was introduced for data-independent acquisition (DIA) for comprehensive protein identification and quantification. Short microflow gradients and the generation of a deep kidney-specific spectral library allowed for high-throughput, comprehensive protein quantification. Upon AKI, the kidney proteome was completely remodeled, and over half of the 3,945 quantified protein groups changed significantly. Downregulated proteins in the injured kidney were involved in energy production, including numerous peroxisomal matrix proteins that function in fatty acid oxidation, such as ACOX1, CAT, EHHADH, ACOT4, ACOT8, and Scp2. Injured mice exhibited severely declined health. The comprehensive and sensitive kidney-specific DIA assays highlighted here feature high-throughput analytical capabilities to achieve deep coverage of the kidney proteome and will serve as useful tools for developing novel therapeutics to remediate kidney function.
Collapse
|
38
|
Wang B, Kim K, Tian M, Kameishi S, Zhuang L, Okano T, Huang Y. Engineered Bone Marrow Stem Cell-Sheets Alleviate Renal Damage in a Rat Chronic Glomerulonephritis Model. Int J Mol Sci 2023; 24:ijms24043711. [PMID: 36835123 PMCID: PMC9959772 DOI: 10.3390/ijms24043711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Although mesenchymal stem cell (MSC)-based regenerative therapy is being developed for the treatment of kidney diseases, cell delivery and engraftment still need to be improved. Cell sheet technology has been developed as a new cell delivery method, to recover cells as a sheet form retaining intrinsic cell adhesion proteins, which promotes its transplantation efficiency to the target tissue. We thus hypothesized that MSC sheets would therapeutically reduce kidney disease with high transplantation efficiency. When the chronic glomerulonephritis was induced by two injections of the anti-Thy 1.1 antibody (OX-7) in rats, the therapeutic efficacy of rat bone marrow stem cell (rBMSC) sheet transplantation was evaluated. The rBMSC-sheets were prepared using the temperature-responsive cell-culture surfaces and transplanted as patches onto the surface of two kidneys of each rat at 24 h after the first injection of OX-7. At 4 weeks, retention of the transplanted MSC-sheets was confirmed, and the animals with MSC-sheets showed significant reductions in proteinuria, glomerular staining for extracellular matrix protein, and renal production of TGFß1, PAI-1, collagen I, and fibronectin. The treatment also ameliorated podocyte and renal tubular injury, as evidenced by a reversal in the reductions of WT-1, podocin, and nephrin and by renal overexpression of KIM-1 and NGAL. Furthermore, the treatment enhanced gene expression of regenerative factors, and IL-10, Bcl-2, and HO-1 mRNA levels, but reduced TSP-1 levels, NF-kB, and NAPDH oxidase production in the kidney. These results strongly support our hypothesis that MSC-sheets facilitated MSC transplantation and function, and effectively retarded progressive renal fibrosis via paracrine actions on anti-cellular inflammation, oxidative stress, and apoptosis and promoted regeneration.
Collapse
Affiliation(s)
- Bin Wang
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health Science, Salt Lake City, UT 84132, USA
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah Health Science, Salt Lake City, UT 84112, USA
| | - Mi Tian
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health Science, Salt Lake City, UT 84132, USA
| | - Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah Health Science, Salt Lake City, UT 84112, USA
| | - Lili Zhuang
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health Science, Salt Lake City, UT 84132, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah Health Science, Salt Lake City, UT 84112, USA
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
- Correspondence: (T.O.); (Y.H.); Tel.: +801-585-0581 (Y.H.); Fax: +801-213-2563 (Y.H.)
| | - Yufeng Huang
- Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah Health Science, Salt Lake City, UT 84132, USA
- Correspondence: (T.O.); (Y.H.); Tel.: +801-585-0581 (Y.H.); Fax: +801-213-2563 (Y.H.)
| |
Collapse
|
39
|
Azırak S. Prevention of nephrotoxicity induced by amikacin: The role of misoprostol, A prostaglandin E1 analogue. Prostaglandins Other Lipid Mediat 2023; 164:106682. [PMID: 36349661 DOI: 10.1016/j.prostaglandins.2022.106682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/05/2022]
Abstract
Amikacin (AK) is an aminoglycoside that is widely used to treat life-threatening Gram-negative infections, especially in intensive care units. Despite its wide clinical indications, AK causes serious side effects such as kidney toxicity. AK was found to lead to tissue damage primarily through apoptosis and oxidative stress. Therefore, it was investigated whether misoprostol (MP), which has antioxidant and antiapoptotic properties, had a beneficial effect on kidney damage caused by AK. It was observed that kidney injury molecule-1 (KIM-1) mRNA, blood urea nitrogen (BUN), creatinine (Cr), NADPH oxidase-4 (NOX-4) and Caspase-3 (CAS-3) levels increased in the AK-treated group in comparison with the control group, while uric acid, albumin, and total protein levels were decreased. In rats that were treated with AK+MP, the levels of KIM-1 mRNA, BUN, Cr, NOX-4 and CAS-3 were significantly decreased in comparison with the AK group, while uric acid, albumin and total protein levels increased. According to the obtained results, MP was found to be quite effective in the protection of kidneys from the toxic effects of AK.
Collapse
Affiliation(s)
- Sebile Azırak
- Vocational School of Health Services, University of Adıyaman, Adıyaman, Turkey.
| |
Collapse
|
40
|
Francis MR, El-Sheakh AR, Suddek GM. Saroglitazar, a dual PPAR-α/γ agonist, alleviates LPS-induced hepatic and renal injury in rats. Int Immunopharmacol 2023; 115:109688. [PMID: 36681027 DOI: 10.1016/j.intimp.2023.109688] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/18/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023]
Abstract
BACKGROUND Lipopolysaccharide (LPS), an endotoxin within gram-negative bacteria, is associated with systemic acute inflammatory response after invading living tissues and results in sepsis. The liver and kidney are both major target organs in sepsis. Septic acute hepatic-renal injury is a serious clinical condition with high risk of morbidity and mortality. Nevertheless, effective treatment is still lacking. AIM This study highlights saroglitazar (SAR), a dual PPAR-α/γ agonist, as a proposed prophylactic drug against LPS-induced hepatic-renal injury. MAIN METHODS Rats were pretreated with SAR (2 and 4 mg/kg/day) for 15 days, while sepsis was induced by LPS injection (10 mg/kg) on day 15 one hour following SAR oral administration. KEY FINDINGS SAR pretreatment could successfully mitigate LPS-induced hepatic-renal injury, evidenced by enhancement of renal and hepatic functions and a decrease of tissue pathological injury. Meanwhile, SAR alleviated LPS-induced oxidative stress; it reduced malondialdehyde (MDA) levels and ameliorated decreased levels of superoxide dismutase (SOD) and glutathione (GSH). LPS-induced elevations in hepatic and renal nuclear factor-kappa B (NF-κB), phosphorylated inhibitor of kappa B alpha (p-IκBα), interferon-beta (IFN-β), and hepatic high mobility group box-1 (HMGB-1) contents were significantly attenuated in SAR-treated groups. SAR showed an advantageous impact against LPS-induced activation of non-canonical inflammasome and pyroptosis via a significant reduction in cysteinyl aspartate-specific proteinase-11 (Caspase-11) and gasdermin D (GSDMD) expressions. Moreover, Nucleotide-Binding Oligomerization Domain (NOD)-Like Receptor Protein 3 (NLRP3) inflammasome activation with concomitant expression and activation of caspase-1 and release of interleukin-1beta (IL-1β) were considerably diminished following SAR pretreatment. SIGNIFICANCE SAR could be considered a prophylactic anti-inflammatory antioxidant drug against LPS-induced liver and kidney injury.
Collapse
Affiliation(s)
- Marina R Francis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Ahmed R El-Sheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
41
|
Rigatto MH, Bergo P, Baldissera G, Beck E, David L, Santoro L, Barros A, Zanin R, Budelon Gonçalves JI, Falci D, Caumo W, Zavascki AP. Melatonin for prevention of acute kidney injury in patients treated with intravenous polymyxin B: a double-blind, placebo-controlled randomized clinical trial. Clin Microbiol Infect 2022; 29:623-628. [PMID: 36586514 DOI: 10.1016/j.cmi.2022.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To evaluate the effect of melatonin versus placebo on the incidence of acute kidney injury (AKI) in patients treated with polymyxin B. METHODS We performed a single-centre, double-blind, randomized clinical trial (NCT03725267) of 30-mg oral melatonin versus placebo for patients treated with intravenous polymyxin B. Patients aged ≥18 years receiving polymyxin B for ≤48 hours were eligible. Melatonin or placebo pills were administered until the end of polymyxin B treatment or for a maximum of 14 days. The main outcome was any level of AKI. RESULTS Eighty-eight patients were randomized: 44 in the melatonin group and 44 in the placebo group. The study ended prematurely because of polymyxin B shortage during the COVID-19 pandemic. The patients' mean age was 63.6 ± 17.3 years, and 60.2% of the patients were men. Forty-six (52.3%, 23 in each group) patients developed AKI during the follow-up period. The incidence rate of AKI was 81.9/1000 and 77.4/1000 patients per day in melatonin and placebo groups, respectively (hazard ratio, 1.09; 95% CI, 0.61-1.94; p 0.78). Renal failure and 30-day mortality were similar between the groups. Moreover, the incidence of AKI was not different in pre-specified sub-groups. DISCUSSION Melatonin initiated in the first 48 hours of therapy did not reduce the incidence of AKI in patients treated with polymyxin B.
Collapse
Affiliation(s)
- Maria H Rigatto
- Medical School and Medical Sciences Post-Graduation Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Infectious Diseases Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Internal Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Pedro Bergo
- Pontifícia Universidade Católica do Rio Grande do Sul Medical School, Porto Alegre, Brazil
| | - Giulia Baldissera
- Infectious Diseases Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eduarda Beck
- Pontifícia Universidade Católica do Rio Grande do Sul Medical School, Porto Alegre, Brazil
| | - Leonardo David
- Pontifícia Universidade Católica do Rio Grande do Sul Medical School, Porto Alegre, Brazil
| | - Lucas Santoro
- Pontifícia Universidade Católica do Rio Grande do Sul Medical School, Porto Alegre, Brazil
| | - Andressa Barros
- Infection Control Service, Hospital São Lucas da PUCRS, Porto Alegre, Brazil
| | - Rafael Zanin
- Pontifícia Universidade Católica do Rio Grande do Sul, Health and Life Sciences School, Porto Alegre, Brazil
| | - João I Budelon Gonçalves
- Pontifícia Universidade Católica do Rio Grande do Sul, Health and Life Sciences School, Porto Alegre, Brazil
| | - Diego Falci
- Infectious Diseases Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul Medical School, Porto Alegre, Brazil; Infection Control Service, Hospital São Lucas da PUCRS, Porto Alegre, Brazil
| | - Wolnei Caumo
- Medical School and Medical Sciences Post-Graduation Program, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Pain and Palliative Care Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Alexandre P Zavascki
- Infectious Diseases Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Internal Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
42
|
Polyphenol-rich açaí seed extract exhibits reno-protective and anti-fibrotic activities in renal tubular cells and mice with kidney failure. Sci Rep 2022; 12:20855. [PMID: 36460743 PMCID: PMC9718837 DOI: 10.1038/s41598-022-24420-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
The main goal of this study was to evaluate the reno-protective effects of a phenolic-rich Açaí seed extract (ASE) in mice with kidney failure. Kidney failure was induced chemically with an adenine-rich diet (0.25% w/w for 4 weeks) in male CD1 Swiss mice. Mice were then provided daily with ASE (at a dose of ~ 350 mg/kg/day) in drinking water for 4 weeks. Adenine mice exhibited renal dysfunction evidenced by increased proteinuria, increased uremia, extensive tubular atrophy and kidney fibrosis associated with overexpression of pro-fibrotic genes (collagen 1a1, transforming growth factor β1, TGF-β1) and markers of tubular injury (such as Kidney injury molecule-1, KIM-1). ASE was able to beneficially counteract all these effects. ASE improved oxidative damage and fibrosis by decreasing carbonylated protein and MDA concentrations, as well as collagen deposition in renal tissue. ASE decreased the expression of TGF-β1 gene and the abundance of protein TGF-β1 in kidneys. It further decreased both expression and urinary excretion of tubular injury biomarkers, e.g., KIM-1 and Neutrophil gelatinase-associated lipocalin. CKD ASE-treated mice exhibited higher polyphenol content and total antioxidant capacity compared to control mice. ASE further prevented the expression of profibrotic genes in HK2 human tubular cells exposed to uremic toxins. Taken together, these findings suggest that ASE exerted potent reno-protective and anti-fibrotic effects through its antioxidant activity and the modulation of the TGF-β1 pathway.
Collapse
|
43
|
Groves AM, Johnston CJ, Beutner GG, Dahlstrom JE, Koina M, O'Reilly MA, Porter G, Brophy PD, Kent AL. Neonatal hypoxic ischemic encephalopathy increases acute kidney injury urinary biomarkers in a rat model. Physiol Rep 2022; 10:e15533. [PMID: 36541220 PMCID: PMC9768655 DOI: 10.14814/phy2.15533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) is associated with acute kidney injury (AKI) in neonates with birth asphyxia. This study aimed to utilize urinary biomarkers to characterize AKI in an established neonatal rat model of HIE. Day 7 Sprague-Dawley rat pups underwent HIE using the Rice-Vannucci model (unilateral carotid ligation followed by 120 mins of 8% oxygen). Controls included no surgery and sham surgery. Weights and urine for biomarkers (NGAL, osteopontin, KIM-1, albumin) were collected the day prior, daily for 3 days post-intervention, and at sacrifice day 14. Kidneys and brains were processed for histology. HIE pups displayed histological evidence of kidney injury including damage to the proximal tubules, consistent with resolving acute tubular necrosis, and had significantly elevated urinary levels of NGAL and albumin compared to sham or controls 1-day post-insult that elevated for 3 days. KIM-1 significantly increased for 2 days post-HIE. HIE did not significantly alter osteopontin levels. Seven days post-start of experiment, controls were 81.2% above starting weight compared to 52.1% in HIE pups. NGAL and albumin levels inversely correlated with body weight following HIE injury. The AKI produced by the Rice-Vannucci HIE model is detectable by urinary biomarkers, which can be used for future studies of treatments to reduce kidney injury.
Collapse
Affiliation(s)
- Angela M. Groves
- Department of PediatricsUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
- Department of Radiation OncologyUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
| | - Carl J. Johnston
- Department of PediatricsUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
| | - Gisela G. Beutner
- Division of CardiologyUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
| | - Jane E. Dahlstrom
- Department of Anatomical Pathology, ACT PathologyCanberra Health ServicesCanberraAustralia
- College of Health and MedicineAustralian National UniversityCanberraAustralia
| | - Mark Koina
- Department of Anatomical Pathology, ACT PathologyCanberra Health ServicesCanberraAustralia
- College of Health and MedicineAustralian National UniversityCanberraAustralia
| | - Michael A. O'Reilly
- Department of PediatricsUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
| | - George Porter
- Division of CardiologyUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
| | - Patrick D. Brophy
- Department of PediatricsUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
- Division of Nephrology, University of Rochester School of Medicine and DentistryGolisano Children's Hospital at University of Rochester Medical CenterNew YorkRochesterUSA
| | - Alison L. Kent
- Department of PediatricsUniversity of Rochester School of Medicine and DentistryNew YorkRochesterUSA
- College of Health and MedicineAustralian National UniversityCanberraAustralia
| |
Collapse
|
44
|
Li L, Yang Z, Lin J, Chen Z, Zhang B, Fu X. Novel Chemiluminescence Immunoassay (CLIA) for the Sensitive Determination of Kidney Injury Molecule-1 in Human Urine. ANAL LETT 2022. [DOI: 10.1080/00032719.2022.2138903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Lanya Li
- Shuyang Hospital of Traditional Chinese Medicine, Suqian, China
| | | | - Jiayuan Lin
- Jiangsu MDK Biotech. Co., Ltd, Suqian, China
| | | | - Bo Zhang
- Jiangsu MDK Biotech. Co., Ltd, Suqian, China
| | - Xiaoling Fu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
45
|
Heart Failure and Cardiorenal Syndrome: A Narrative Review on Pathophysiology, Diagnostic and Therapeutic Regimens-From a Cardiologist's View. J Clin Med 2022; 11:jcm11237041. [PMID: 36498617 PMCID: PMC9741317 DOI: 10.3390/jcm11237041] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022] Open
Abstract
In cardiorenal syndrome (CRS), heart failure and renal failure are pathophysiologically closely intertwined by the reciprocal relationship between cardiac and renal injury. Type 1 CRS is most common and associated with acute heart failure. A preexistent chronic kidney disease (CKD) is common and contributes to acute kidney injury (AKI) in CRS type 1 patients (acute cardiorenal syndrome). The remaining CRS types are found in patients with chronic heart failure (type 2), acute and chronic kidney diseases (types 3 and 4), and systemic diseases that affect both the heart and the kidney (type 5). Establishing the diagnosis of CRS requires various tools based on the type of CRS, including non-invasive imaging modalities such as TTE, CT, and MRI, adjuvant volume measurement techniques, invasive hemodynamic monitoring, and biomarkers. Albuminuria and Cystatin C (CysC) are biomarkers of glomerular filtration and integrity in CRS and have a prognostic impact. Comprehensive "all-in-one" magnetic resonance imaging (MRI) approaches, including cardiac magnetic resonance imaging (CMR) combined with functional MRI of the kidneys and with brain MRI are proposed for CRS. Hospitalizations due to CRS and mortality are high. Timely diagnosis and initiation of effective adequate therapy, as well as multidisciplinary care, are pertinent for the improvement of quality of life and survival. In addition to the standard pharmacological heart failure medication, including SGLT2 inhibitors (SGLT2i), renal aspects must be strongly considered in the context of CRS, including control of the volume overload (diuretics) with special caution on diuretic resistance. Devices involved in the improvement of myocardial function (e.g., cardiac resynchronization treatment in left bundle branch block, mechanical circulatory support in advanced heart failure) have also shown beneficial effects on renal function.
Collapse
|
46
|
Seyahi NS, Ozcan SG. Application of New Acute Kidney Injury Biomarkers. Biomark Med 2022. [DOI: 10.2174/9789815040463122010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Kidney-related biomarkers can provide structural and functional information
about different parts of the nephron. These biomarkers can be used to evaluate
glomerular, tubular, or interstitial injury, inflammation, or repair, and glomerular or
tubular function. Furthermore, biomarkers can improve the acute kidney injury
diagnosis in various clinical conditions, including acute interstitial nephritis, acute
tubular injury, hepatorenal and cardiorenal syndrome, ischemic and nephrotoxic acute
kidney injury, and drug-induced acute kidney injury. Biomarkers might be used as an
additional precision medicine tool in managing patients with acute kidney injury; they
can help with clinical decision-making and impact patient outcomes. In this chapter, we
reviewed the utility of biomarkers used in acute kidney injury.
Collapse
Affiliation(s)
- Nurhan Seyahi Seyahi
- Department of Nephrology, Cerrahpasa Medical Faculty, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Seyda Gul Ozcan
- Department of Internal Medicine, Cerrahpasa Medical Faculty, Istanbul University -
Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
47
|
Heinzl MW, Resl M, Klammer C, Fellinger P, Schinagl L, Obendorf F, Feldbauer R, Pohlhammer J, Wagner T, Egger M, Dieplinger B, Clodi M. SUBCLINICAL KIDNEY INJURY IS CAUSED BY A MODERATE SINGLE INFLAMMATORY EVENT. Shock 2022; 58:14-19. [PMID: 35616594 PMCID: PMC9415208 DOI: 10.1097/shk.0000000000001942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 04/14/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Background: Current means of diagnosis of acute kidney injury (AKI) based on serum creatinine have poor sensitivity and may miss possible therapeutic windows in subclinical kidney injury, especially in septic AKI. Kidney injury molecule-1 (KIM-1) may be a valuable biomarker to improve diagnostic algorithms for AKI. The understanding of septic AKI is still insufficient, and knowledge about KIM-1 kinetics in inflammation is scarce. The aim of this study was to investigate the possible effect of lipopolysaccharide (LPS) on KIM-1 as a marker of structural kidney injury in healthy volunteers. Methods: A single-blinded, placebo-controlled cross-over study using the human endotoxin model (LPS administration) was performed in 10 healthy men. Kidney injury molecule-1 and serum creatinine were measured repetitively for 48 hours. Results: We observed a significant elevation of serum KIM-1 levels after the administration of LPS ( P < 0.001). Furthermore, LPS caused a significant elevation of serum creatinine at an early time point ( P = 0.013) as compared with placebo. Conclusion: Even a relatively small inflammatory stimulus is sufficient to cause subclinical structural kidney injury with elevated KIM-1 and serum creatinine in healthy volunteers. This outlines the insufficiency of the current diagnostic approach regarding AKI and the urgency to develop novel diagnostic algorithms including markers of kidney injury. Clinical Trial Registration:www.clinicaltrials.gov . Unique identifier: NCT03392701 (August 1, 2018).
Collapse
Affiliation(s)
- Matthias Wolfgang Heinzl
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
- Institute for Cardiovascular and Metabolic Research, Johannes Kepler Universität Linz, Linz, Austria
| | - Michael Resl
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
- Institute for Cardiovascular and Metabolic Research, Johannes Kepler Universität Linz, Linz, Austria
| | - Carmen Klammer
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
- Institute for Cardiovascular and Metabolic Research, Johannes Kepler Universität Linz, Linz, Austria
| | - Paul Fellinger
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lukas Schinagl
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
| | - Florian Obendorf
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
| | - Roland Feldbauer
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
| | - Johannes Pohlhammer
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
| | - Thomas Wagner
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
| | - Margot Egger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
| | - Martin Clodi
- Department of Internal Medicine, Konventhospital Barmherzige Brueder Linz (St. John of God Hospital Linz), Linz, Austria
- Institute for Cardiovascular and Metabolic Research, Johannes Kepler Universität Linz, Linz, Austria
| |
Collapse
|
48
|
Zdanowicz A, Urban S, Ponikowska B, Iwanek G, Zymliński R, Ponikowski P, Biegus J. Novel Biomarkers of Renal Dysfunction and Congestion in Heart Failure. J Pers Med 2022; 12:jpm12060898. [PMID: 35743683 PMCID: PMC9224642 DOI: 10.3390/jpm12060898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/15/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Heart failure is a major public health problem and, despite the constantly emerging, new, effective treatments, it remains a leading cause of morbidity and mortality. Reliable tools for early diagnosis and risk stratification are crucial in the management of HF. This explains a growing interest in the development of new biomarkers related to various pathophysiological mechanisms of HF. In the course of this review, we focused on the markers of congestion and renal dysfunction in terms of their interference with cardiovascular homeostasis. Congestion is a hallmark feature of heart failure, contributing to symptoms, morbidity, and hospitalizations of patients with HF and has, therefore, become a therapeutic target in AHF. On the other hand, impaired renal function by altering the volume status contributes to the development and progression of HF and serves as a marker of an adverse clinical outcome. Early detection of congestion and an adequate assessment of renal status are essential for the prompt administration of patient-tailored therapy. This review provides an insight into recent advances in the field of HF biomarkers that could be potentially implemented in diagnosis and risk stratification of patients with HF.
Collapse
Affiliation(s)
- Agata Zdanowicz
- Institute of Heart Diseases, Medical University, 50-556 Wroclaw, Poland; (A.Z.); (G.I.); (R.Z.); (P.P.); (J.B.)
| | - Szymon Urban
- Institute of Heart Diseases, Medical University, 50-556 Wroclaw, Poland; (A.Z.); (G.I.); (R.Z.); (P.P.); (J.B.)
- Correspondence: ; Tel.: +48-71-733-11-12
| | - Barbara Ponikowska
- Student Scientific Organization, Institute of Heart Diseases, Medical University, 50-556 Wroclaw, Poland;
| | - Gracjan Iwanek
- Institute of Heart Diseases, Medical University, 50-556 Wroclaw, Poland; (A.Z.); (G.I.); (R.Z.); (P.P.); (J.B.)
| | - Robert Zymliński
- Institute of Heart Diseases, Medical University, 50-556 Wroclaw, Poland; (A.Z.); (G.I.); (R.Z.); (P.P.); (J.B.)
| | - Piotr Ponikowski
- Institute of Heart Diseases, Medical University, 50-556 Wroclaw, Poland; (A.Z.); (G.I.); (R.Z.); (P.P.); (J.B.)
| | - Jan Biegus
- Institute of Heart Diseases, Medical University, 50-556 Wroclaw, Poland; (A.Z.); (G.I.); (R.Z.); (P.P.); (J.B.)
| |
Collapse
|
49
|
Tang Y, Liu T, Cai Q, Zhao M. The Effects of Febuxostat on Urine NGAL and Urine KIM-1 in Patients with Hyperuricemia. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:6028611. [PMID: 35419185 PMCID: PMC9001067 DOI: 10.1155/2022/6028611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 11/24/2022]
Abstract
Retrospective analysis of the effects of febuxostat on urine neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) in patients with hyperuricemia was performed. From January 2018 to June 2018, there were 45 patients with asymptomatic hyperuricemia in the outpatient or inpatient of Changzhou Second People's Hospital, which were divided into the febuxostat group (25 cases) and the control group (20 cases). We collected the patients' baseline indicators and testing indicators after three months of treatment, including blood urea nitrogen, blood creatinine, blood uric acid, urine microalbumin, urine NGAL, urine KIM-1, and other indicators. The subjects in both groups were given lifestyle intervention, instructed to drink more water, and given a low-purine diet. The patients in the febuxostat group took febuxostat 40 mg/D or 80 mg/D. We used SPSS 25.0 statistical software for statistical analysis. Baseline indexes between the febuxostat group and the control group and indexes after treatment between two groups were both performed by independent sample t-test, and paired t-test was used for self-comparison between the groups before and after treatment. There was no significant difference in age, sex, body mass index (BMI), urea nitrogen, creatinine, uric acid, urine microalbumin/creatinine, urine NGAL/creatinine, and urine KIM-1/creatinine between the two groups before treatment (P > 0.05). Compared with before treatment, after 3 months of intervention, the levels of serum uric acid, urine microalbumin/creatinine, urine NGAL/creatinine, and urine KIM-1/creatinine were significantly decreased in the febuxostat group (P < 0.05), while the changes of blood urea nitrogen, serum creatinine, and epidermal growth factor receptor (eGFR) were not statistically significant (P > 0.05). After 3 months of intervention, the control group had no significant changes in blood urea nitrogen, creatinine, eGFR, uric acid, microalbumin/creatinine, urine NGAL/creatinine, and urine KIM-1/creatinine (P > 0.05). After 3 months of intervention, compared with the control group, the serum uric acid, microalbumin/creatinine, urine NGAL/creatinine, and urine KIM-1/creatinine were significantly decreased in the febuxostat group (P < 0.05), but there was no significant difference in blood urea nitrogen, creatinine, and eGFR (P > 0.05). Febuxostat can reduce urine NGAL/creatinine and urine KIM-1/creatinine levels in patients with hyperuricemia and has the protective effects on renal tubular injury caused by hyperuricemia, which can provide evidences for the early prevention and treatment of asymptomatic hyperuricemia.
Collapse
Affiliation(s)
- Yushang Tang
- Department of Nephrology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Tongqiang Liu
- Department of Nephrology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Qiuping Cai
- Department of Nephrology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Minwen Zhao
- Department of Nephrology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| |
Collapse
|
50
|
Urinary Activin A: A Novel Biomarker for Human Acute Kidney Injury. Diagnostics (Basel) 2022; 12:diagnostics12030661. [PMID: 35328214 PMCID: PMC8947696 DOI: 10.3390/diagnostics12030661] [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: 01/13/2022] [Revised: 02/23/2022] [Accepted: 03/07/2022] [Indexed: 02/01/2023] Open
Abstract
Activin is a multifunctional cytokine belonging to the transforming growth factor (TGF)-β superfamily that regulates the growth and differentiation of cells in various organs. We previously reported that activin A, which is absent in normal kidneys, was significantly increased in the ischemic kidney, and that the blockade of activin action by follistatin, an activin antagonist, significantly enhanced tubular regeneration after renal ischemia, suggesting that activin A acts as an endogenous inhibitor of tubular repair after kidney injury in rodents. However, the role of activin A in human acute kidney injury (AKI) remains unclear. In this analysis, we measured serum and urinary activin A in human AKI (n = 39) and tested if activin A might serve as a biomarker for AKI. Urinary activin A, which was undetectable in healthy controls, was significantly increased in AKI (0.0 ± 0.0 vs. 173.4 ± 58.8 pg/mL, p < 0.05). The urinary activin A level in patients with AKI stage 3, was significantly higher than that in patients with AKI stages 1 and 2. Patients who required renal replacement therapy (RRT) had a significantly higher urinary activin A level than patients who did not require RRT. Urinary activin A might be a useful non-invasive biomarker for the severity of AKI.
Collapse
|