1
|
Chang M, Shi X, Yang B, Li P, Zhang Y, Zhang Q, Zhang Y. Modified Huangqi Chifeng decoction alleviates podocyte injury on rat with experimental membranous nephropathy. Ren Fail 2025; 47:2459896. [PMID: 39972601 PMCID: PMC11843643 DOI: 10.1080/0886022x.2025.2459896] [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: 09/16/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVE This study aims to investigate the therapeutic effects of modified Huangqi Chifeng decoction (MHCD) on proteinuria in membranous nephropathy (MN) and its potential protective effects on podocytes. Furthermore, we explored whether these effects are associated with the inhibition of the nuclear factor kappa-B (NF-κB) pathway. METHODS Passive Heymann nephritis (PHN) rat model was applied with a single tail vein injection of sheep anti-rat Fx1A serum (0.4 ml/100g). All rats were divided into four groups: normal group, PHN group, benazepril group (10 mg/kg), and MHCD group (12.5 g/kg), and were treated for 6 weeks. 24-hour urine protein levels and serum biochemical parameters were measured. Optical microscopy and transmission electron microscopy were performed to assess pathological changes in renal tissues. Additionally, the expression levels of IgG, C5b-9, nephrin, podocin, Wilms' tumor gene 1 (WT-1), and NF-κB p65 were evaluated. RESULTS PHN rats exhibited progressive proteinuria over time. However, MHCD treatment significantly reduced levels of proteinuria and triglyceride, while increased levels of albumin. Moreover, MHCD alleviated pathological damage in renal tissues, and reduced the expression of IgG and membrane attack complex (C5b-9). Immunohistochemistry analysis revealed that MHCD increased the expression of nephrin, podocin, and WT-1. Western blot analysis showed that MHCD increased the expression of nephrin and podocin while inhibiting the activation of NF-κB p65. CONCLUSIONS Our findings indicate that MHCD exert reno-protective effects in the experimental rat model of MN by alleviating podocyte damage and inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Meiying Chang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Deparment of Nephrology, The First Hospital of Tsinghua University, Beijing, China
| | - Xiujie Shi
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Deparment of Nephrology, The First Hospital of Tsinghua University, Beijing, China
| | - Bin Yang
- Department of Pathology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Li
- Experimental Research Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yifan Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Qi Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Jiao B, An C, Du H, Tran M, Yang D, Zhao Y, Wang P, Hu Z, Zhou D, Wang Y. Genetic deficiency or pharmacological inhibition of cGAS-STING signalling suppresses kidney inflammation and fibrosis. Br J Pharmacol 2025; 182:1741-1762. [PMID: 39833988 DOI: 10.1111/bph.17412] [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: 10/24/2023] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Chronic kidney disease (CKD) is characterised by inflammation, which can lead to tubular atrophy and fibrosis. The molecular mechanisms are not well understood. In this study, we investigated the functional role of the cyclic GMP-AMP synthase (cGAS)- stimulator of interferon genes (STING) signalling in renal inflammation and fibrosis. EXPERIMENTAL APPROACH Mice with global cGAS deficiency or global or myeloid cell-specific STING deficiency or wild-type mice treated with RU.521, a selective cGAS inhibitor, were used to examine the role of cGAS-STING signalling in renal inflammation and fibrosis in a preclinical model of obstructive nephropathy in vivo. Bone marrow-derived macrophages were used to determine whether tubular epithelial cell-derived DNA can activate cGAS-STING signalling in vitro. KEY RESULTS Following obstructive injury, cGAS-STING signalling was activated in the kidneys during the development of renal fibrosis. Mice with deficiency of cGAS or STING exhibited significantly less macrophage proinflammatory activation, myofibroblast formation, total collagen deposition, and extracellular matrix (ECM) protein production in the kidneys following obstructive injury. Pharmacological inhibition of cGAS with RU.521 reduced macrophage proinflammatory activation, suppressed myofibroblast formation, and attenuated kidney fibrosis following obstructive injury. Mechanistically, cGAS-STING signalling in macrophages is activated by double-stranded DNA released from damaged tubular epithelial cells, which induces inflammatory responses. CONCLUSIONS AND IMPLICATIONS Our study identifies the cGAS-STING signalling pathway as a critical regulator of macrophage proinflammatory activation during the development of renal fibrosis. Therefore, inhibition of cGAS-STING signalling may represent a novel therapeutic strategy for CKD.
Collapse
Affiliation(s)
- Baihai Jiao
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Changlong An
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Hao Du
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Melanie Tran
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Duomeng Yang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Yuqi Zhao
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Monrovia, California, USA
| | - Penghua Wang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Zhaoyong Hu
- Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut, USA
- Renal Section, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
3
|
Guarino S, Calcaterra V, Di Sessa A, Labati L, Marrapodi MM, Grandone A, Zanfardino A, Zuccotti G, Iafusco D, Miraglia Del Giudice E, Marzuillo P. Sensitivity to thyroid hormones in children developing acute kidney injury at the onset of type 1 diabetes mellitus. BMC Med 2025; 23:123. [PMID: 40011875 DOI: 10.1186/s12916-025-03936-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Thyroid hormone (TH) sensitivity at type 1 diabetes mellitus (T1DM) onset and its connection with acute kidney injury (AKI) has not been investigated. We aimed to evaluate changes in TH sensitivity in children with and without AKI at T1DM onset and to assess the role of euthyroid sick syndrome (ESS) in this relationship. METHODS We included 161 children with new-onset T1DM and followed them until renal function normalized. The free triiodothyronine (FT3)/free thyroxine (FT4) ratio was used to assess peripheral TH sensitivity, while the TSH index (TSHI), thyrotroph T4 resistance index (TT4RI), thyrotroph T3 resistance index (TT3RI), Thyroid Feedback Quantile-based Index (TFQI), and parametric TFQI (PTFQI) were used for central sensitivity. RESULTS Patients with AKI exhibited greater weight loss, higher serum ketones, creatinine, corrected sodium, and glycated hemoglobin, but lower bicarbonate and estimated glomerular filtration rate compared to those without AKI. Logistic regression showed that the odds of AKI increased by 11.5-fold for each unit decrease in TFQI, 4.0-fold per unit decrease in PTFQI, and 1.7-fold per unit decrease in TSHI, adjusting for age and gender. After adjusting for age, gender, and ESS, the odds for AKI significantly increased (4.8-fold for each 1-unit decrease) only for TFQI. CONCLUSIONS AKI at the onset of T1DM has a dual effect on TH. It reduces peripheral sensitivity while increasing central sensitivity. This effect appears to be largely driven by ESS, with the exception of the association between AKI and TFQI, which remains independent of ESS.
Collapse
Affiliation(s)
- Stefano Guarino
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Valeria Calcaterra
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100, Pavia, Italy
- Pediatric Department, Buzzi Children's Hospital, 20154, Milan, Italy
| | - Anna Di Sessa
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Labati
- Pediatric Department, Buzzi Children's Hospital, 20154, Milan, Italy
- Department of Biomedical and Clinical Science, University of Milan, 20157, Milan, Italy
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Anna Grandone
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Angela Zanfardino
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children's Hospital, 20154, Milan, Italy
- Department of Biomedical and Clinical Science, University of Milan, 20157, Milan, Italy
| | - Dario Iafusco
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Emanuele Miraglia Del Giudice
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Pierluigi Marzuillo
- Department of Woman, Child and of General and Specialized Surgery, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
4
|
Lim SW, Jo WY, Park HP. A low preoperative platelet-to-white blood cell ratio is associated with acute kidney injury following cerebral aneurysm treatment in South Korea. Acute Crit Care 2025; 40:59-68. [PMID: 39978949 PMCID: PMC11924346 DOI: 10.4266/acc.003120] [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: 08/01/2024] [Accepted: 12/03/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Inflammation is involved in the pathophysiology of postoperative acute kidney injury (AKI). We investigated whether preoperative platelet-to-white blood cell ratio (PWR), a novel serum biomarker of systemic inflammation, was associated with postoperative AKI following cerebral aneurysm treatment. We also compared the discrimination power of preoperative PWR with those of other preoperative systemic inflammatory indices in predicting postoperative AKI. METHODS Perioperative data including preoperative systemic inflammatory indices and cerebral aneurysm-related variables were retrospectively analyzed in 4,429 cerebral aneurysm patients undergoing surgical clipping or endovascular coiling. Based on the cutoff value of preoperative PWR, patients were divided into the high PWR (≥39.04, n=1,924) and low PWR (<39.04, n=2,505) groups. After propensity score matching (PSM), 1,168 patients in each group were included in the data analysis. AKI was defined according to the Kidney Disease Improving Global Outcomes guidelines. RESULTS Postoperative AKI occurred more frequently in the low PWR group than in the high PWR group before PSM (45 [1.8%] vs. 7 [0.4%], P<0.001) and after (17 [1.5%] vs. 5 [0.4%], P=0.016). A low preoperative PWR was predictive of postoperative AKI before PSM (odds ratio [95% CI], 3.93 [1.74-8.87]; P<0.001) and after (3.44 [1.26-9.34], P=0.016). Preoperative PWR showed the highest area under the curve for postoperative AKI (0.713 [0.644-0.782], P<0.001), followed by preoperative platelet-to-neutrophil ratio (0.694 [0.619-0.769], P<0.001), neutrophil percentage-to-albumin ratio (0.671 [0.592-0.750], P<0.001), white blood cell-to-hemoglobin ratio (0.665 [0.579-0.750], P<0.001), neutrophil-to-lymphocyte ratio (0.648 [0.569-0.728], P<0.001), and systemic inflammatory index (0.615 [0.532-0.698], P=0.004). CONCLUSIONS A low preoperative PWR was associated with postoperative AKI following cerebral aneurysm treatment.
Collapse
Affiliation(s)
- Seung-Woon Lim
- Department of Anesthesiology and Pain Medicine, Chung-Ang University Hospital, Seoul, Korea
| | - Woo-Young Jo
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hee-Pyoung Park
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
5
|
Li X, Xu R, Zhang D, Cai J, Zhou H, Song T, Wang X, Kong Q, Li L, Liu Z, He Z, Tang Z, Tan J, Zhang J. Baicalin: a potential therapeutic agent for acute kidney injury and renal fibrosis. Front Pharmacol 2025; 16:1511083. [PMID: 39911847 PMCID: PMC11795133 DOI: 10.3389/fphar.2025.1511083] [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: 10/14/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Acute kidney injury (AKI) is a common critical clinical disease that is linked to significant morbidity, recurrence, and mortality. It is characterized by a fast and prolonged loss in renal function arising from numerous etiologies and pathogenic pathways. Renal fibrosis, defined as the excessive accumulation of collagen and proliferation of fibroblasts within renal tissues, contributes to the structural damage and functional decline of the kidneys, playing a pivotal role in the advancement of Chronic Kidney Disease (CKD). Until now, while continuous renal replacement therapy (CRRT) has been utilized in the management of severe AKI, there remains a dearth of effective targeted therapies for AKI stemming from diverse etiologies. Similarly, the identification of specific biomarkers and pharmacological targets for the treatment of renal fibrosis remains a challenge. Baicalin, a naturally occurring compound classified within the flavonoid group and commonly found in the Chinese herb Scutellaria baicalensis, has shown a range of pharmacological characteristics, such as antioxidant, anti-inflammatory, antifibrotic, antitumor and antiviral effects, as evidenced by research studies. Research shows that Baicalin has potential in treating kidney diseases like AKI and renal fibrosis. This review aims to summarize Baicalin's progress in these areas, including its molecular mechanism, application in treatment, and absorption, distribution, metabolism, and excretion. Baicalin's therapeutic effects are achieved through various pathways, including antioxidant, anti-inflammatory, antifibrosis, and regulation of apoptosis and cell proliferation. Besides, we also hope this review may give some enlightenment for treating AKI and renal fibrosis in clinical practice.
Collapse
Affiliation(s)
- Xiaoming Li
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Rui Xu
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Dan Zhang
- Zunyi Medical University Library Administrative Office, Zunyi, China
| | - Ji Cai
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - He Zhou
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Qinghong Kong
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, China
| | - Liujin Li
- Department of Otolaryngology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhaohui Liu
- Department of Otolaryngology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| | - Zhengzhen Tang
- Department of Pediatrics, The First People’s Hospital of Zunyi, Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Tan
- Department of Histology and Embryology, Zunyi Medical University, Zunyi, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| |
Collapse
|
6
|
Hariri G, Legrand M. New drugs for acute kidney injury. JOURNAL OF INTENSIVE MEDICINE 2025; 5:3-11. [PMID: 39872831 PMCID: PMC11763585 DOI: 10.1016/j.jointm.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 01/30/2025]
Abstract
Acute kidney injury (AKI) presents a significant challenge in the management of critically ill patients, as it is associated with increased mortality, prolonged hospital stays, and increased healthcare costs. In certain conditions, such as during sepsis or after cardiac surgery, AKI is one of the most frequent complications, affecting 30%-50% of patients. Over time, even after the resolution of AKI, it can evolve into chronic kidney disease, a leading global cause of mortality, and cardiovascular complications. Despite significant improvement in the care of critically ill patients over the past two decades, the incidence of AKI remains stable, and novel approaches aiming at reducing its occurrence or improving AKI outcomes are still mostly lacking. However, recent insights into the pathophysiology of AKI within critical care settings have shed light on new pathways for both prevention and treatment, providing various new therapeutic targets aimed to mitigating kidney injury. These advancements highlight the intricate and multifaceted nature of the mechanisms underlying AKI, which could explain the challenge of identifying an effective treatment. Among these targets, modulation of the inflammatory responses and the cellular metabolism, hemodynamic regulation and enhancement of cellular repair mechanisms, have emerged as promising options. These multifaceted approaches offer renewed hope for limiting the incidence and severity of AKI in critically ill patients. Several ongoing clinical trials are evaluating the efficacy of these different strategies and we are facing an exiting time with multiple therapeutic interventions being tested to prevent or treat AKI. In this review, we aim to provide a summary of the new drugs evaluated for preventing or treating AKI in critical care and surgical settings.
Collapse
Affiliation(s)
- Geoffroy Hariri
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, UCSF, San Francisco, CA, USA
- Sorbonne Université, GRC 29, Assistance Publique-Hôpitaux de Paris (AP-HP), DMU DREAM, Département d'anesthésie et réanimation, Institut de Cardiologie, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Matthieu Legrand
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, UCSF, San Francisco, CA, USA
- Investigation Network Initiative Cardiovascular and Renal Clinical Trialist Network, Nancy, France
| |
Collapse
|
7
|
Gubernatorova EO, Samsonov MY, Drutskaya MS, Lebedeva S, Bukhanova D, Materenchuk M, Mutig K. Targeting inerleukin-6 for renoprotection. Front Immunol 2024; 15:1502299. [PMID: 39723211 PMCID: PMC11668664 DOI: 10.3389/fimmu.2024.1502299] [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: 09/26/2024] [Accepted: 11/08/2024] [Indexed: 12/28/2024] Open
Abstract
Sterile inflammation has been increasingly recognized as a hallmark of non-infectious kidney diseases. Induction of pro-inflammatory cytokines in injured kidney tissue promotes infiltration of immune cells serving to clear cell debris and facilitate tissue repair. However, excessive or prolonged inflammatory response has been associated with immune-mediated tissue damage, nephron loss, and development of renal fibrosis. Interleukin 6 (IL-6) is a cytokine with pleiotropic effects including a major role in inflammation. IL-6 signals either via membrane-bound (classic signaling) or soluble receptor forms (trans-signaling) thus affecting distinct cell types and eliciting various metabolic, cytoprotective, or pro-inflammatory reactions. Antibodies neutralizing IL-6 or its receptor have been developed for therapy of autoimmune and chronic non-renal inflammatory diseases. Small molecule inhibitors of Janus kinases acting downstream of the IL-6 receptor, as well as recombinant soluble glycoprotein 130 variants suppressing the IL-6 trans-signaling add to the available therapeutic options. Animal data and accumulating clinical experience strongly suggest that suppression of IL-6 signaling pathways bears therapeutic potential in acute and chronic kidney diseases. The present work analyses the renoprotective potential of clinically relevant IL-6 signaling inhibitors in acute kidney injury, chronic kidney disease, and kidney transplantation with focus on current achievements and future prospects.
Collapse
Affiliation(s)
- Ekaterina O. Gubernatorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Marina S. Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Russia
| | - Svetlana Lebedeva
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Medical Elementology, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | | | - Maria Materenchuk
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kerim Mutig
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
8
|
do Carmo JM, Hall JE, Furukawa LNS, Woronik V, Dai X, Ladnier E, Wang Z, Omoto ACM, Mouton A, Li X, Luna-Suarez EM, da Silva AA. Chronic central nervous system leptin administration attenuates kidney dysfunction and injury in a model of ischemia/reperfusion-induced acute kidney injury. Am J Physiol Renal Physiol 2024; 327:F957-F966. [PMID: 39361725 PMCID: PMC11687842 DOI: 10.1152/ajprenal.00158.2024] [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: 05/28/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024] Open
Abstract
In the present study, we examined whether chronic intracerebroventricular (ICV) leptin administration protects against ischemia/reperfusion (I/R)-induced acute kidney injury (AKI). Twelve-week-old male rats were implanted with an ICV cannula into the right lateral ventricle, and 8-10 days after surgery, leptin (0.021 µg/h, n = 8) or saline vehicle (0.5 µL/h, n = 8) was infused via osmotic minipump connected to the ICV cannula for 12 days. On day 8 of leptin or vehicle infusion, rats were submitted to unilateral ischemia/reperfusion (UIR) by clamping the left pedicle for 30 min. To control for leptin-induced reductions in food intake, the vehicle-treated group was pair-fed (UIR-PF) to match the same amount of food consumed by leptin-treated (UIR-Leptin) rats. On the 12th day of leptin or vehicle infusion (fourth day after AKI), single-left kidney glomerular filtration rate (GFR) was measured, blood samples were collected to quantify white blood cells, and kidneys were collected for histological assessment of injury. UIR-Leptin-treated rats showed reduced right and left kidney weights (right: 1,040 ± 24 vs. 1,281 ± 36 mg; left: 1,127 ± 71 vs. 1,707 ± 45 mg, for UIR-Leptin and UIR-PF, respectively). ICV leptin infusion improved GFR (0.50 ± 0.06 vs. 0.13 ± 0.03 mL/min/g kidney wt) and reduced kidney injury scores. ICV leptin treatment also attenuated the reduction in circulating adiponectin levels that was observed in UIR-PF rats and increased the circulating white blood cells count compared with UIR-PF rats (16.3 ± 1.3 vs. 9.8 ± 0.6 k/µL). Therefore, we show that leptin, via its actions on the central nervous system, confers significant protection against major kidney dysfunction and injury in a model of ischemia/reperfusion-induced AKI.NEW & NOTEWORTHY A major new finding of this study is that chronic activation of leptin receptors in the CNS markedly attenuates acute kidney injury and protects against severe renal dysfunction after ischemia/reperfusion, independently of leptin's anorexic effects.
Collapse
Affiliation(s)
- Jussara M do Carmo
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - John E Hall
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Luzia N S Furukawa
- Laboratory of Renal Pathophysiology, Department of Internal Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Viktoria Woronik
- Laboratory of Renal Pathophysiology, Department of Internal Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Xuemei Dai
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Emily Ladnier
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Zhen Wang
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Ana C M Omoto
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alan Mouton
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Xuan Li
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Emilio M Luna-Suarez
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
9
|
Fontecha-Barriuso M, Villar-Gomez N, Guerrero-Mauvecin J, Martinez-Moreno JM, Carrasco S, Martin-Sanchez D, Rodríguez-Laguna M, Gómez MJ, Sanchez-Niño MD, Ruiz-Ortega M, Ortiz A, Sanz AB. Runt-related transcription factor 1 (RUNX1) is a mediator of acute kidney injury. J Pathol 2024; 264:396-410. [PMID: 39472111 DOI: 10.1002/path.6355] [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: 02/07/2024] [Revised: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 11/09/2024]
Abstract
Treatment for acute kidney injury (AKI) is suboptimal. A better understanding of the pathogenesis of AKI may lead to new therapeutic approaches. Kidney transcriptomics of folic acid-induced AKI (FA-AKI) in mice identified Runx1 as the most upregulated RUNX family gene. We then examined the expression of RUNX1 in FA-AKI, in bacterial lipopolysaccharide (LPS)-induced cytokine storm-AKI (CS-AKI), and in human AKI. In cultured mouse tubule cells, we explored the expression and role of RUNX1 in response to the cytokine TWEAK or LPS. A chemical inhibitor of RUNX1 (Ro5-3335) was used in animal models of AKI to test its potential as a therapeutic target. RUNX1 overexpression in FA-AKI was validated at the mRNA and protein levels and localized mainly to tubule cell nuclei. CS-AKI also upregulated kidney RUNX1. Increased tubule and interstitial RUNX1 expression were also observed in human AKI. In cultured mouse tubule cells, the pro-inflammatory cytokine TWEAK and LPS increased RUNX1 and IL-6 expression. Mechanistically, RUNX1 bound to the Il6 gene promoter and RUNX1 targeting with the chemical inhibitor Ro5-3335, or a specific small interfering RNA (siRNA), prevented the TWEAK- and LPS-induced upregulation of IL6 through a RUNX1/NFκB1 p50 pathway. In vivo, preventive Ro5-3335 improved kidney function and reduced inflammation in FA-AKI and CS-AKI. However, Ro5-3335 administration after the insult only improved kidney function in CS-AKI. Kidney transcriptomics identified inflammatory genes and transcription factor mRNAs such as Yap1 and Trp53 as key targets of Ro5-3335 in CS-AKI. In conclusion, RUNX1 contributes to AKI by driving the expression of genes involved in inflammation and represents a novel therapeutic target in AKI. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Miguel Fontecha-Barriuso
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Natalia Villar-Gomez
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Juan Guerrero-Mauvecin
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Julio M Martinez-Moreno
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
| | - Susana Carrasco
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
| | - Diego Martin-Sanchez
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
| | | | - Manuel J Gómez
- Unidad de Bioinformatica, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María D Sanchez-Niño
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
- RICORS2040, Madrid, Spain
- Department of Pharmacology, Universidad Autonoma de Madrid, Madrid, Spain
| | - Marta Ruiz-Ortega
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
- RICORS2040, Madrid, Spain
- Department of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
- RICORS2040, Madrid, Spain
- Department of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
- IRSIN, Madrid, Spain
| | - Ana B Sanz
- Laboratorio de Nefrología Experimental, Instituto de Investigación Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid, Madrid, Spain
- RICORS2040, Madrid, Spain
| |
Collapse
|
10
|
Payasi A, Yadav MK, Chaudhary S, Aggarwal A. Evaluating nephrotoxicity reduction in a novel polymyxin B formulation: insights from a 3D kidney-on-a-chip model. Antimicrob Agents Chemother 2024; 68:e0021924. [PMID: 39225483 PMCID: PMC11459911 DOI: 10.1128/aac.00219-24] [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: 02/09/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to assess the nephrotoxicity associated with VRP-034 (novel formulation of polymyxin B [PMB]) compared to marketed PMB in a three-dimensional (3D) kidney-on-a-chip model. To model the human kidney proximal tubule for analysis, tubular structures were established using 23 triple-channel chips seeded with RPTEC/hTERT1 cells. These cells were exposed to VRP-034 or PMB at seven concentrations (1-200 µM) over 12, 24, and 48 h. A suite of novel kidney injury biomarkers, cell health, and inflammatory markers were quantitatively assessed in the effluent. Additionally, caspase and cytochrome C levels were measured, and cell viability was evaluated using calcein AM and ethidium homodimer-1 (EthD-1). Exposure to marketed PMB resulted in significantly elevated levels (P < 0.05) of four key biomarkers (KIM-1, cystatin C, clusterin, and OPN) compared to VRP-034, particularly at clinically relevant concentrations of ≥10 µM. At 25 µM, all biomarkers demonstrated a significant increase (P < 0.05) with marketed PMB exposure compared to VRP-034. Inflammatory markers (interleukin-6 and interleukin-8) increased significantly (P < 0.05) with marketed PMB at concentrations of ≥5 µM, relative to VRP-034. VRP-034 displayed superior cell health outcomes, exhibiting lower lactate dehydrogenase release, while ATP levels remained comparable. Morphological analysis revealed that marketed PMB induced more severe damage, disrupting tubular integrity. Both treatments activated cytochrome C, caspase-3, caspase-8, caspase-9, and caspase-12 in a concentration-dependent manner; however, caspase activation was significantly reduced (P < 0.05) with VRP-034. This study demonstrates that VRP-034 significantly reduces nephrotoxicity compared to marketed PMB within a 3D microphysiological system, suggesting its potential to enable the use of full therapeutic doses of PMB with an improved safety profile, addressing the need for less nephrotoxic polymyxin antibiotics.
Collapse
Affiliation(s)
- Anurag Payasi
- Department of Cell Culture, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| | - Manoj Kumar Yadav
- Department of Cell Culture, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| | | | - Anmol Aggarwal
- Department of Pipeline Strategy, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| |
Collapse
|
11
|
Zhang DL, Chen S, Xu JM, Na-Lin, Wu HY, Zhou JM, Chen ZP, Huang XR, Wei LX, Liu DX. The value of tissue quantitative diffusion analysis of ultrasound elastography in the diagnosis of early-stage chronic kidney disease. BMC Nephrol 2024; 25:328. [PMID: 39354395 PMCID: PMC11446147 DOI: 10.1186/s12882-024-03762-0] [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: 11/24/2023] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
PURPOSE To explore the value of tissue quantitative diffusion analysis of ultrasound elastography in the diagnosis of early-stage chronic kidney disease (CKD). METHODS The observation group comprised 54 patients with early-stage CKD treated at Fuzhou No 7 Hospital, and the control group consisted of 40 healthy individuals who underwent physical examinations at the same hospital. The renal parenchyma of the participants were examined using ultrasonography, color Doppler ultrasonography, and tissue quantitative diffusion analysis of ultrasound elastography. Renal dimensions (diameter, thickness, and renal parenchyma thickness), interlobar artery blood flow parameters, and 11 elastic characteristic values were analyzed and compared between the two groups. The area under the receiver-operating characteristic (ROC) curve, cut-off values, sensitivity, and specificity were calculated using the ROC curve analysis. RESULTS There were no significant differences in the blood flow parameters of the interlobar artery and the dimensions of renal meridians between the two groups. In the observation group, the mean (MEAN) decreased, while the blue area ratio and skewness, increased, compared to the control group (p < 0.05). In addition, the ROC curve revealed that the blue area ratio, MEAN, and skewness had significant diagnostic value (the area under the curve > 0.7). Notably, the best cut-off value of the MEAN was found to be 106, indicating that a MEAN value less than 106 represented early-stage CKD. Also, this cutoff value had a sensitivity of 80% and a specificity of 81%. CONCLUSION Tissue quantitative diffusion analysis of ultrasound elastography can quantitatively evaluate renal parenchymal damage in early-stage CKD using quantitative diffusion parameters, with the MEAN parameter, having a cutoff of 106, being particularly effective. This parameter and cutoff value offer a valuable tool for the early detection and diagnosis of CKD, potentially improving patient outcomes through earlier intervention. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Dan-Ling Zhang
- Department of Ultrasonography, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China.
- Department of Ultrasonography, Fuzhou No 7 Hospital, Fuzhou, Fujian, 350005, China.
| | - Sheng Chen
- Department of Ultrasonography, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Jia-Ming Xu
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Na-Lin
- Department of Ultrasonography, Fuzhou No 7 Hospital, Fuzhou, Fujian, 350005, China
| | - Hai-Yan Wu
- Department of Ultrasonography, Fuzhou No 7 Hospital, Fuzhou, Fujian, 350005, China
| | - Jin-Mei Zhou
- Department of Ultrasonography, Fuzhou No 7 Hospital, Fuzhou, Fujian, 350005, China
| | - Zhao-Ping Chen
- Department of Ultrasonography, Fuzhou No 7 Hospital, Fuzhou, Fujian, 350005, China
| | - Xu-Ri Huang
- Department of Physician, Fuzhou No 7 Hospital, Fuzhou, Fujian, 350005, China
| | - Li-Xin Wei
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Dai-Xiang Liu
- Department of Physician, Fuzhou No 7 Hospital, Fuzhou, Fujian, 350005, China
| |
Collapse
|
12
|
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
|
13
|
ALRashdi BM, Massoud D, Rashwan HK, Mahgoub S, Abuelezz NZ, Nasr AM, Kassab RB, Amin HK. The Protecting Role of Black Seed Oil and Its Nano-Formulation in LPS-Induced Acute Kidney Injury in Mice: Evaluation of Oxidative Stress, Biochemical & Molecular Parameters. J Inflamm Res 2024; 17:4747-4763. [PMID: 39051058 PMCID: PMC11268590 DOI: 10.2147/jir.s463369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Background Acute kidney injury (AKI) is a medical concern that is accompanied by the rapid deterioration of kidney function. It can be triggered by lipopolysaccharide (LPS) of gram-negative bacteria as it activates a complicated immune response, resulting in widespread inflammation and potential organ dysfunction. Black seed oil (BSO) is rich in beneficial constituents and has been widely used owing to its nutritional advantages. Purpose This research is aimed to investigate the potential protective effects of BSO and its nano-formulation on AKI induced by LPS. It also aimed to compare their anti-inflammatory activity with indomethacin, a known synthetic anti-inflammatory drug. Materials and Methods Forty-eight mice were placed randomly into 8 groups. A single intraperitoneal (i.p.) injection of 2.5 mg/kg B.W. of LPS was used to trigger inflammation, and pretreatment with BSO and its nano-formulation was at 0.2 mL/kg/day for 14 consecutive days. Indomethacin was used as a reference drug and its efficacy was tested alone or in combination with BSO at lower doses. Renal function was assessed using urea, creatinine, neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1). Also, oxidative and inflammatory markers were assessed by measuring levels of reduced glutathione (GSH), nitric oxide (NO), cyclooxygenase-2 (COX-2), tumor necrosis factor alpha (TNF-α), and toll-like receptor-4 (TLR-4). Histopathological examination of the kidney tissues was also performed. Results The study showed that BSO and its nano-formulation had anti-inflammatory effects comparable to or better than those of indomethacin. They greatly decreased the oxidative stress and inflammatory markers induced by LPS. Their protective effect against pathological alterations in kidney tissues was significantly noticed. Conclusion BSO and its nano-formulation could be used as nephroprotective and anti-inflammatory supplements.
Collapse
Affiliation(s)
- Barakat M ALRashdi
- Department of Biology, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Diaa Massoud
- Department of Biology, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Hager K Rashwan
- Department of Biochemistry, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, Egypt
| | - Shahenda Mahgoub
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Nermeen Z Abuelezz
- Department of Biochemistry, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, Egypt
| | - Ali M Nasr
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, Port Said, 42526, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Galala University, New Galala, 43713, Egypt
| | - Rami B Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Hatem K Amin
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
14
|
Tang J, Wu C, Zhong Z. Group-Based Trajectory Modeling of Fluid Balance in Elderly Patients with Acute Ischemic Stroke: Analysis from Multicenter ICUs. Neurol Ther 2024; 13:749-761. [PMID: 38635141 PMCID: PMC11136917 DOI: 10.1007/s40120-024-00612-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
INTRODUCTION Acute ischemic stroke (AIS) significantly contributes to severe disability and mortality among the elderly. This study aims to explore the association between longitudinal fluid balance (FB) trajectories and clinical outcomes in elderly patients with AIS. Our hypothesis posits the existence of multiple latent trajectories of FB in patients with AIS during the initial 7 days following ICU admission. METHODS Patients (age ≥ 65 years) with AIS and continuous FB records were identified from two large databases. Group-based trajectory modeling identified latent groups with similar 7-day FB trajectories. Subsequently, multivariable logistic and quasi-Poisson regression were employed to evaluate the relationship between trajectory groups and outcomes. Additionally, nonlinear associations between maximum fluid overload (FO) and outcomes were analyzed using restricted cubic spline models. To further validate our findings, subgroup and sensitivity analysis were conducted. RESULTS A total of 1146 eligible patients were included in this study, revealing three trajectory patterns were identified: low FB (84.8%), decreasing FB (7.2%), and high FB (7.9%). High FB emerged as an independent risk factor for in-hospital mortality. Compared with those without FO, patients with FO had a 1.57-fold increased risk of hospital mortality (adjusted odd ratio (OR) 1.57, 95% confidence interval (CI) 1.08-2.27), 2.37-fold increased risk of adverse kidney event (adjusted OR 2.37, 95% CI 1.56-3.59), and 1.33-fold increased risk of prolonged ICU stay (adjusted incidence rate ratio (IRR) 1.33, 95% CI 1.19-1.48). The risk of hospital mortality and adverse kidney event increased linearly with rising maximum FO (P for non-linearity = 0.263 and 0.563, respectively). CONCLUSION Daily FB trajectories were associated with adverse outcomes in elderly patients with AIS. Regular assessment of daily fluid status and restriction of FO are crucial for the recovery of critically ill patients.
Collapse
Affiliation(s)
- Jia Tang
- Graduate School of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Changdong Wu
- Xinjiang Emergency Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91, Tian-Chi Road, Tianshan District, Urumqi, 830001, Xinjiang, China.
| | - Zhenguang Zhong
- Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
15
|
Wang B, Wang Y, Wang J, Jin C, Zhou R, Guo J, Zhang H, Wang M. Multiparametric Magnetic Resonance Investigations on Acute and Long-Term Kidney Injury. J Magn Reson Imaging 2024; 59:43-57. [PMID: 37246343 DOI: 10.1002/jmri.28784] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/30/2023] Open
Abstract
Acute kidney injury (AKI) is a frequent complication of critical illness and carries a significant risk of short- and long-term mortality. The prediction of the progression of AKI to long-term injury has been difficult for renal disease treatment. Radiologists are keen for the early detection of transition from AKI to long-term kidney injury, which would help in the preventive measures. The lack of established methods for early detection of long-term kidney injury underscores the pressing needs of advanced imaging technology that reveals microscopic tissue alterations during the progression of AKI. Fueled by recent advances in data acquisition and post-processing methods of magnetic resonance imaging (MRI), multiparametric MRI is showing great potential as a diagnostic tool for many kidney diseases. Multiparametric MRI studies offer a precious opportunity for real-time noninvasive monitoring of pathological development and progression of AKI to long-term injury. It provides insight into renal vasculature and function (arterial spin labeling, intravoxel incoherent motion), tissue oxygenation (blood oxygen level-dependent), tissue injury and fibrosis (diffusion tensor imaging, diffusion kurtosis imaging, T1 and T2 mapping, quantitative susceptibility mapping). The multiparametric MRI approach is highly promising but the longitudinal investigation on the transition of AKI to irreversible long-term impairment is largely ignored. Further optimization and implementation of renal MR methods in clinical practice will enhance our comprehension of not only AKI but chronic kidney diseases. Novel imaging biomarkers for microscopic renal tissue alterations could be discovered and benefit the preventative interventions. This review explores recent MRI applications on acute and long-term kidney injury while addressing lingering challenges, with emphasis on the potential value of the development of multiparametric MRI for renal imaging on clinical systems. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Bin Wang
- Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongfang Wang
- Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Jinxia Guo
- GE Healthcare, MR Research China, Beijing, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Min Wang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Barrera-Chimal J, Henley N, Grant MP, Cenatus S, Geraldes P, Pichette V, Gerarduzzi C. Tungsten toxicity on kidney tubular epithelial cells induces renal inflammation and M1-macrophage polarization. Cell Biol Toxicol 2023; 39:3061-3075. [PMID: 37368165 DOI: 10.1007/s10565-023-09817-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023]
Abstract
Tungsten is widely used in medical, industrial, and military applications. The environmental exposure to tungsten has increased over the past several years, and few studies have addressed its potential toxicity. In this study, we evaluated the effects of chronic oral tungsten exposure (100 ppm) on renal inflammation in male mice. We found that 30- or 90-day tungsten exposure led to the accumulation of LAMP1-positive lysosomes in renal tubular epithelial cells. In addition, the kidneys of mice exposed to tungsten showed interstitial infiltration of leukocytes, myeloid cells, and macrophages together with increased levels of proinflammatory cytokines and p50/p65-NFkB subunits. In proximal tubule epithelial cells (HK-2) in vitro, tungsten induced a similar inflammatory status characterized by increased mRNA levels of CSF1, IL34, CXCL2, and CXCL10 and NFkB activation. Moreover, tungsten exposure reduced HK-2 cell viability and enhanced reactive oxygen species generation. Conditioned media from HK-2 cells treated with tungsten induced an M1-proinflammatory polarization of RAW macrophages as evidenced by increased levels of iNOS and interleukin-6 and decreased levels of the M2-antiinflammatory marker CD206. These effects were not observed when RAW cells were exposed to conditioned media from HK-2 cells treated with tungsten and supplemented with the antioxidant N-acetylcysteine (NAC). Similarly, direct tungsten exposure induced M1-proinflammatory polarization of RAW cells that was prevented by NAC co-treatment. Altogether, our data suggest that prolonged tungsten exposure leads to oxidative injury in the kidney ultimately leading to chronic renal inflammation characterized by a proinflammatory status in kidney tubular epithelial cells and immune cell infiltration.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montreal, Quebec, Canada
| | - Nathalie Henley
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montreal, Quebec, Canada
| | - Michael Philip Grant
- Department of Orthopaedics, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Schrodinger Cenatus
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montreal, Quebec, Canada
| | - Pedro Geraldes
- Research Center, Centre Hospitalier, Université de Sherbrooke, Quebec, Canada
| | - Vincent Pichette
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montreal, Quebec, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Casimiro Gerarduzzi
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montreal, Quebec, Canada.
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
Price N, Wood AF. Acute kidney injury in the critical care setting. Nurs Stand 2023; 38:45-50. [PMID: 37458070 DOI: 10.7748/ns.2023.e12063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 07/18/2023]
Abstract
Acute kidney injury is a sudden reduction in renal function which impairs the kidneys' ability to maintain fluid, electrolyte and acid-base balance. The syndrome often develops secondary to severe illness and is associated with a significant increase in morbidity and mortality rate in critically ill patients. This article gives an overview of the pathophysiology and aetiology of acute kidney injury, as well as the associated complications and clinical diagnostic signs. The authors also describe some common causes of the syndrome in critically ill patients, specifically sepsis, liver failure and cardiac failure, and discuss patient management in the critical care setting, with a focus on haemodynamic support and continuous renal replacement therapy.
Collapse
Affiliation(s)
- Natasha Price
- division of nursing and paramedic science, school of health sciences, Queen Margaret University, Edinburgh, Scotland
| | - Alison Fiona Wood
- programme lead for independent prescribing, division of nursing and paramedic science, school of health sciences, Queen Margaret University, Edinburgh, Scotland
| |
Collapse
|
18
|
Calleri A, Alessandria C. Renal damage in Hepatorenal Syndrome: A still unsolved issue. Clin Res Hepatol Gastroenterol 2023; 47:102178. [PMID: 37453679 DOI: 10.1016/j.clinre.2023.102178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/02/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Acute kidney injury (AKI) is a common complication of cirrhosis, burdened by high morbidity and mortality rates and progression to chronic kidney disease. Hepatorenal syndrome (HRS) is a peculiar type of functional AKI observed in cirrhotic patients with ascites. HRS diagnosis is still clinical, once pre-renal azotemia and intrinsic kidney damage have been excluded by applying well-established and internationally adopted criteria. HRS is considered reversible because of the absence of intrinsic renal damage. However, HRS reversibility has been questioned, due to the lack of response to treatment with vasoconstrictors plus albumin in a relevant percentage of patients and to the persistence of renal dysfunction in HRS patients who underwent liver transplantation (LT). Indeed, LT is the only ultimate treatment, as it solves both liver failure and portal hypertension. Thus, the presence of renal damage in HRS can be hypothesized. In this scenario, neutrophil gelatinase-associated lipocalin (NGAL), one of the most promising biomarkers, may help in characterizing the type of renal injury, distinguishing between HRS and acute tubular necrosis. This review gathers the available evidence in favor and against the presence of structural lesions in HRS in terms of either renal histology and urinary biomarkers with a particular focus on NGAL. The ability to properly characterize which component of renal dysfunction prevails - functional rather than structural - entails a relevant clinical impact for the treatment of these patients, both in terms of medical therapy and liver vs. combined liver-kidney transplantation.
Collapse
Affiliation(s)
- Alberto Calleri
- Division of Gastroenterology and Hepatology, Città della Salute e della Scienza Hospital, University of Turin, Italy
| | - Carlo Alessandria
- Division of Gastroenterology and Hepatology, Città della Salute e della Scienza Hospital, University of Turin, Italy.
| |
Collapse
|
19
|
Baer PC, Neuhoff AK, Schubert R. microRNA Expression of Renal Proximal Tubular Epithelial Cells and Their Extracellular Vesicles in an Inflammatory Microenvironment In Vitro. Int J Mol Sci 2023; 24:11069. [PMID: 37446246 DOI: 10.3390/ijms241311069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Renal proximal tubular epithelial cells (PTCs) are central players during renal inflammation. In response to inflammatory signals, PTCs not only self-express altered mRNAs, microRNAs (miRNAs), proteins, and lipids, but also release altered extracellular vesicles (EVs). These EVs also carry inflammation-specific cargo molecules and are key players in cell-cell-communication. Understanding the precise molecular and cellular mechanisms that lead to inflammation in the kidney is the most important way to identify early targets for the prevention or treatment of acute kidney injury. Therefore, highly purified human PTCs were used as an in vitro model to study the cellular response to an inflammatory microenvironment. A cytokine-induced inflammatory system was established to analyze different miRNA expression in cells and their EVs. In detail, we characterized the altered miR expression of PTCs and their released EVs during induced inflammation and showed that 12 miRNAs were significantly regulated in PTCs (6 upregulated and 6 downregulated) and 9 miRNAs in EVs (8 upregulated and 1 downregulated). We also showed that only three of the miRNAs were found to overlap between cells and EVs. As shown by the KEGG pathway analysis, these three miRNAs (miR-146a-5p, miR-147b, and miR-155-5p) are functionally involved in the regulation of the Toll-like receptor signaling pathway and significantly correlated with the inflammatory mediators IL6 and ICAM1 released by stimulated PTCs. Especially with regard to a possible clinical use of miRs as new biomarkers, an accurate characterization of the miR expression altered during inflammatory processes is of enormous importance.
Collapse
Affiliation(s)
- Patrick C Baer
- Department of Internal Medicine 4, Nephrology, University Hospital, Goethe-University, 60596 Frankfurt/M., Germany
| | - Ann-Kathrin Neuhoff
- Division of Allergology, Pneumology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, Goethe-University, 60596 Frankfurt/M., Germany
| | - Ralf Schubert
- Division of Allergology, Pneumology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, Goethe-University, 60596 Frankfurt/M., Germany
| |
Collapse
|
20
|
Deshpande K, Lange KR, Stone WB, Yohn C, Schlesinger N, Kagan L, Auguste AJ, Firestein BL, Brunetti L. The influence of SARS-CoV-2 infection on expression of drug-metabolizing enzymes and transporters in a hACE2 murine model. Pharmacol Res Perspect 2023; 11:e01071. [PMID: 37133236 PMCID: PMC10155506 DOI: 10.1002/prp2.1071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 05/04/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting Coronavirus disease 2019 emerged in late 2019 and is responsible for significant morbidity and mortality worldwide. A hallmark of severe COVID-19 is exaggerated systemic inflammation, regarded as a "cytokine storm," which contributes to the damage of various organs, primarily the lungs. The inflammation associated with some viral illnesses is known to alter the expression of drug-metabolizing enzymes and transporters. These alterations can lead to modifications in drug exposure and the processing of various endogenous compounds. Here, we provide evidence to support changes in the mitochondrial ribonucleic acid expression of a subset of drug transporters (84 transporters) in the liver, kidneys, and lungs and metabolizing enzymes (84 enzymes) in the liver in a humanized angiotensin-converting enzyme 2 receptor mouse model. Specifically, three drug transporters (Abca3, Slc7a8, Tap1) and the pro-inflammatory cytokine IL-6 were upregulated in the lungs of SARS-CoV-2 infected mice. We also found significant downregulation of drug transporters responsible for the movement of xenobiotics in the liver and kidney. Additionally, expression of cytochrome P-450 2f2 which is known to metabolize some pulmonary toxicants, was significantly decreased in the liver of infected mice. The significance of these findings requires further exploration. Our results suggest that further research should emphasize altered drug disposition when investigating therapeutic compounds, whether re-purposed or new chemical entities, in other animal models and ultimately in individuals infected with SARS-CoV-2. Moreover, the influence and impact of these changes on the processing of endogenous compounds also require further investigation.
Collapse
Affiliation(s)
- Kiran Deshpande
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Center of Excellence in Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Keith R. Lange
- Department of Cell Biology and Neuroscience, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - William B. Stone
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science InstituteVirginia Polytechnic Institute and State UniversityVirginiaUSA
| | - Christine Yohn
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Center of Excellence in Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Naomi Schlesinger
- Division of RheumatologyDepartment of Medicine, Rutgers Robert Wood Johnson Medical SchoolNew BrunswickNew JerseyUSA
| | - Leonid Kagan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Center of Excellence in Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Albert J. Auguste
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science InstituteVirginia Polytechnic Institute and State UniversityVirginiaUSA
- Center for Emerging, Zoonotic, and Arthropod‐borne PathogensVirginia Polytechnic Institute and State UniversityBlacksburgVirginiaUSA
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Luigi Brunetti
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Center of Excellence in Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| |
Collapse
|
21
|
Abdelmageed MM, Kefaloyianni E, Arthanarisami A, Komaru Y, Atkinson JJ, Herrlich A. TNF or EGFR inhibition equally block AKI-to-CKD transition: opportunities for etanercept treatment. Nephrol Dial Transplant 2023; 38:1139-1150. [PMID: 36269313 PMCID: PMC10157768 DOI: 10.1093/ndt/gfac290] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Inflammation is a key driver of the transition of acute kidney injury to progressive fibrosis and chronic kidney disease (AKI-to-CKD transition). Blocking a-disintegrin-and-metalloprotease-17 (ADAM17)-dependent ectodomain shedding, in particular of epidermal growth factor receptor (EGFR) ligands and of the type 1 inflammatory cytokine tumor necrosis factor (TNF), reduces pro-inflammatory and pro-fibrotic responses after ischemic AKI or unilateral ureteral obstruction (UUO), a classical fibrosis model. Metalloprotease or EGFR inhibition show significant undesirable side effects in humans. In retrospective studies anti-TNF biologics reduce the incidence and progression of CKD in humans. Whether TNF has a role in AKI-to-CKD transition and how TNF inhibition compares to EGFR inhibition is largely unknown. METHODS Mice were subjected to bilateral renal ischemia-reperfusion injury or unilateral ureteral obstruction. Kidneys were analyzed by histology, immunohistochemistry, qPCR, western blot, mass cytometry, scRNA sequencing, and cytokine profiling. RESULTS Here we show that TNF or EGFR inhibition reduce AKI-to-CKD transition and fibrosis equally by about 25%, while combination has no additional effect. EGFR inhibition reduced kidney TNF expression by about 50% largely by reducing accumulation of TNF expressing immune cells in the kidney early after AKI, while TNF inhibition did not affect EGFR activation or immune cell accumulation. Using scRNAseq data we show that TNF is predominantly expressed by immune cells in AKI but not in proximal tubule cells (PTC), and PTC-TNF knockout did not affect AKI-to-CKD transition in UUO. Thus, the anti-inflammatory and anti-fibrotic effects of the anti-TNF biologic etanercept in AKI-to-CKD transition rely on blocking TNF that is released from immune cells recruited or accumulating in response to PTC-EGFR signals. CONCLUSION Short-term anti-TNF biologics during or after AKI could be helpful in the prevention of AKI-to-CKD transition.
Collapse
Affiliation(s)
- Mai M Abdelmageed
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Eirini Kefaloyianni
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Akshayakeerthi Arthanarisami
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Yohei Komaru
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| | - Jeffrey J Atkinson
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Pulmonary and Critical Care Medicine
| | - Andreas Herrlich
- Washington University School of Medicine in Saint Louis, Department of Medicine, St. Louis, MO, USA
- Division of Nephrology
| |
Collapse
|
22
|
Florens N, Kasam RK, Rudman-Melnick V, Lin SC, Prasad V, Molkentin JD. Interleukin-33 Mediates Cardiomyopathy After Acute Kidney Injury by Signaling to Cardiomyocytes. Circulation 2023; 147:746-758. [PMID: 36695175 PMCID: PMC9992318 DOI: 10.1161/circulationaha.122.063014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/19/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Acute kidney injury (AKI) is a short-term life-threatening condition that, if survived, can lead to renal insufficiency and development of chronic kidney disease. The pathogenesis of AKI and chronic kidney disease involves direct effects on the heart and the development of hypertrophy and cardiomyopathy. METHODS We used mouse models of ischemia/reperfusion AKI and unilateral ureteral obstruction to investigate the role of IL-33 (interleukin-33) and its receptor-encoding gene Il1rl1 (also called ST2L [suppression of tumorigenicity 2]) in cardiac remodeling after AKI. Mice with cell type-specific genetic disruption of the IL-33/ST2L axis were used, and IL-33 monoclonal antibody, adeno-associated virus encoding IL-33 or ST2L, and recombinant IL-33, as well. RESULTS Mice deficient in Il33 were refractory to cardiomyopathy associated with 2 models of kidney injury. Treatment of mice with monoclonal IL-33 antibody also protected the heart after AKI. Moreover, overexpression of IL-33 or injection of recombinant IL-33 induced cardiac hypertrophy or cardiomyopathy, but not in mice lacking Il1rl1. AKI-induced cardiomyopathy was also reduced in mice with cardiac myocyte-specific deletion of Il1rl1 but not in endothelial cell- or fibroblast-specific deletion of Il1rl1. Last, overexpression of the ST2L receptor in cardiac myocytes recapitulated induction of cardiac hypertrophy. CONCLUSIONS These results indicate that IL-33 released from the kidney during AKI underlies cardiorenal syndrome by directly signaling to cardiac myocytes, suggesting that antagonism of IL-33/ST2 axis would be cardioprotective in patients with kidney disease.
Collapse
Affiliation(s)
- Nans Florens
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Rajesh K. Kasam
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Valeria Rudman-Melnick
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Suh-Chin Lin
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffery D. Molkentin
- Department of Pediatrics, Cincinnati Children’s Hospital and the University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
23
|
Prostaglandin transporter PGT as a new pharmacological target in the prevention of inflammatory cytokine-induced injury in renal proximal tubular HK-2 cells. Life Sci 2023; 313:121260. [PMID: 36473541 DOI: 10.1016/j.lfs.2022.121260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/20/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
|
24
|
Zhang W, Liu L, Xiao X, Zhou H, Peng Z, Wang W, Huang L, Xie Y, Xu H, Tao L, Nie W, Yuan X, Liu F, Yuan Q. Identification of common molecular signatures of SARS-CoV-2 infection and its influence on acute kidney injury and chronic kidney disease. Front Immunol 2023; 14:961642. [PMID: 37026010 PMCID: PMC10070855 DOI: 10.3389/fimmu.2023.961642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the main cause of COVID-19, causing hundreds of millions of confirmed cases and more than 18.2 million deaths worldwide. Acute kidney injury (AKI) is a common complication of COVID-19 that leads to an increase in mortality, especially in intensive care unit (ICU) settings, and chronic kidney disease (CKD) is a high risk factor for COVID-19 and its related mortality. However, the underlying molecular mechanisms among AKI, CKD, and COVID-19 are unclear. Therefore, transcriptome analysis was performed to examine common pathways and molecular biomarkers for AKI, CKD, and COVID-19 in an attempt to understand the association of SARS-CoV-2 infection with AKI and CKD. Three RNA-seq datasets (GSE147507, GSE1563, and GSE66494) from the GEO database were used to detect differentially expressed genes (DEGs) for COVID-19 with AKI and CKD to search for shared pathways and candidate targets. A total of 17 common DEGs were confirmed, and their biological functions and signaling pathways were characterized by enrichment analysis. MAPK signaling, the structural pathway of interleukin 1 (IL-1), and the Toll-like receptor pathway appear to be involved in the occurrence of these diseases. Hub genes identified from the protein-protein interaction (PPI) network, including DUSP6, BHLHE40, RASGRP1, and TAB2, are potential therapeutic targets in COVID-19 with AKI and CKD. Common genes and pathways may play pathogenic roles in these three diseases mainly through the activation of immune inflammation. Networks of transcription factor (TF)-gene, miRNA-gene, and gene-disease interactions from the datasets were also constructed, and key gene regulators influencing the progression of these three diseases were further identified among the DEGs. Moreover, new drug targets were predicted based on these common DEGs, and molecular docking and molecular dynamics (MD) simulations were performed. Finally, a diagnostic model of COVID-19 was established based on these common DEGs. Taken together, the molecular and signaling pathways identified in this study may be related to the mechanisms by which SARS-CoV-2 infection affects renal function. These findings are significant for the effective treatment of COVID-19 in patients with kidney diseases.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Leping Liu
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Hongshan Zhou
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
| | - Wei Wang
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
| | - Ling Huang
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
| | - Hui Xu
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
| | - Wannian Nie
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Xiangning Yuan
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
| | - Fang Liu
- Health Management Center, Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Fang Liu, ; Qiongjing Yuan,
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
- Organ Fibrosis Key Lab of Hunan Province, Central South University, Changsha, China
- National Clinical Medical Research Center for Geriatric Diseases, Xiangya Hospital of Central South University, Changsha, China
- Research Center for Medical Metabolomics, Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Fang Liu, ; Qiongjing Yuan,
| |
Collapse
|
25
|
Tain YL, Hsu CN. Cardiovascular Risks of Hypertension: Lessons from Children with Chronic Kidney Disease. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1650. [PMID: 36360378 PMCID: PMC9688449 DOI: 10.3390/children9111650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023]
Abstract
Hypertension is the most common complication of chronic kidney disease (CKD) in children, having a strong association with subsequential cardiovascular disease (CVD). In pediatric CKD, a considerable percentage of children with hypertension are undiagnosed or undertreated. Prior research has evaluated structural and functional markers of subclinical CVD and biomarkers in adults with CKD, while ideal biomarkers in pediatrics are still insufficiently studied. The ultimate goal of this review is to summarize what is currently known about state of hypertension, cardiovascular risk factors, and potential CVD markers/biomarkers in children with pre-dialysis CKD. We discuss omics-related biomarkers and the pathophysiologic processes of endothelial dysfunction, kidney injury, oxidative stress and inflammation that are classified by specific biomarkers. Moreover, we illustrate the existing challenges and highlight the paucity of pediatric CKD research to evaluate these CVD biomarkers for future clinical pediatric practice. Thus, achieving clinical utility of CVD biomarkers for use in pediatric CKD remains a significant challenge requiring additional efforts.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
26
|
Feng YL, Yang Y, Chen H. Small molecules as a source for acute kidney injury therapy. Pharmacol Ther 2022; 237:108169. [DOI: 10.1016/j.pharmthera.2022.108169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
|
27
|
Li B, Lin F, Xia Y, Ye Z, Yan X, Song B, Yuan T, Li L, Zhou X, Yu W, Cheng F. The Intersection of Acute Kidney Injury and Non-Coding RNAs: Inflammation. Front Physiol 2022; 13:923239. [PMID: 35755446 PMCID: PMC9218900 DOI: 10.3389/fphys.2022.923239] [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: 04/19/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022] Open
Abstract
Acute renal injury (AKI) is a complex clinical syndrome, involving a series of pathophysiological processes, in which inflammation plays a key role. Identification and verification of gene signatures associated with inflammatory onset and progression are imperative for understanding the molecular mechanisms involved in AKI pathogenesis. Non-coding RNAs (ncRNAs), involved in epigenetic modifications of inflammatory responses, are associated with the aberrant expression of inflammation-related genes in AKI. However, its regulatory role in gene expression involves precise transcriptional regulation mechanisms which have not been fully elucidated in the complex and volatile inflammatory response of AKI. In this study, we systematically review current research on the intrinsic molecular mechanisms of ncRNAs that regulate the inflammatory response in AKI. We aim to provide potential research directions and strategies for developing ncRNA-targeted gene therapies as an intervention for the inflammatory damage in AKI.
Collapse
Affiliation(s)
- Bojun Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuqi Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zehua Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinzhou Yan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baofeng Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tianhui Yuan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Zhu Z, Hu J, Chen Z, Feng J, Yang X, Liang W, Ding G. Transition of acute kidney injury to chronic kidney disease: role of metabolic reprogramming. Metabolism 2022; 131:155194. [PMID: 35346693 DOI: 10.1016/j.metabol.2022.155194] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is a global public health concern associated with high morbidity and mortality. Although advances in medical management have improved the in-hospital mortality of severe AKI patients, the renal prognosis for AKI patients in the later period is not encouraging. Recent epidemiological investigations have indicated that AKI significantly increases the risk for the development of chronic kidney disease (CKD) and end-stage renal disease (ESRD) in the future, further contributing to the economic burden on health care systems. The transition of AKI to CKD is complex and often involves multiple mechanisms. Recent studies have suggested that renal tubular epithelial cells (TECs) are more prone to metabolic reprogramming during AKI, in which the metabolic process in the TECs shifts from fatty acid β-oxidation (FAO) to glycolysis due to hypoxia, mitochondrial dysfunction, and disordered nutrient-sensing pathways. This change is a double-edged role. On the one hand, enhanced glycolysis acts as a compensation pathway for ATP production; on the other hand, long-term shut down of FAO and enhanced glycolysis lead to inflammation, lipid accumulation, and fibrosis, contributing to the transition of AKI to CKD. This review discusses developments and therapies focused on the metabolic reprogramming of TECs during AKI, and the emerging questions in this evolving field.
Collapse
Affiliation(s)
- Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Xueyan Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
29
|
Kumar MNVR. Urolithin A Nanoparticle Therapy for Cisplatin-Induced Acute Kidney Injury. Nephron Clin Pract 2022; 147:3-5. [PMID: 35472845 PMCID: PMC9755071 DOI: 10.1159/000524509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022] Open
Abstract
Cisplatin continues to be one of the frontline cytotoxic drugs. However, cisplatin-induced acute kidney injury (AKI) remains a major unmet medical need without any approved pharmacological interventions. The involvement of reactive oxygen species generation and activation of inflammatory and apoptotic pathways in the pathogenesis of cisplatin-induced AKI prompts the use of natural anti-inflammatory compounds. In this context, resolution of inflammation using natural antioxidant and anti-inflammatory such as urolithin A (UA) could prove beneficial. In the end, testing such combinations in models to eliminate the possibility that UA stimulates tumor growth or compromises the potency of cisplatin could prove useful for clinical translation of adjuvant therapies.
Collapse
Affiliation(s)
- M N V Ravi Kumar
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama, USA
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama, USA
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama, USA
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
30
|
Ross L. Acute Kidney Injury in Dogs and Cats. Vet Clin North Am Small Anim Pract 2022; 52:659-672. [DOI: 10.1016/j.cvsm.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
31
|
Rodas L, Barnadas E, Pereira A, Castrejon N, Saurina A, Calls J, Calzada Y, Madrid Á, Blasco M, Poch E, García-Herrera A, Quintana LF. The Density of Renal Lymphatics Correlates With Clinical Outcomes in IgA Nephropathy. Kidney Int Rep 2022; 7:823-830. [PMID: 35497787 PMCID: PMC9039908 DOI: 10.1016/j.ekir.2021.12.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 11/06/2022] Open
Abstract
Introduction IgA nephropathy (IgAN) is the most common primary glomerulonephritis (GN) worldwide. The disease course fluctuates, and the most important challenge is the considerable variation in the time lag between diagnosis and the development of a hard clinical end point, such as end-stage kidney disease (ESKD). The reaction of renal tissue to damage resembles the common wound-healing response. One part of this repair in IgAN is the expansion of lymphatic vessels known as lymphangiogenesis. The aim of this work was to establish the prognostic value of the density of lymphatic vessels in the renal biopsy at the time of diagnosis, for predicting the risk of ESKD in a Spanish cohort of patients with IgAN. Methods We performed a retrospective multicenter study of 76 patients with IgAN. The end point of the study was progression to ESKD. The morphometric analysis of lymphatic vessels was performed on tissue sections stained with antipodoplanin antibody. Results Density of lymphatic vessels was significantly higher in patients with IgAN with mesangial hypercellularity >50%, segmental sclerosis, higher degrees of interstitial fibrosis, and tubular atrophy. Patients with more lymphatic vessels had significantly higher values of proteinuria and lower estimated glomerular filtration rate (eGFR). A density of lymphatic vessels ≥8 per mm2 was associated with a significantly higher rate of progression to ESKD at 3 years from biopsy. After adjustment for the International IgAN prediction score, at the multivariate logistic regression, high density of lymphatic vessels (≥8 per mm2) remained significantly associated with a higher rate of early progression to ESKD. Conclusion This study contributes to the understanding of the natural history of the progression to ESKD in patients with IgAN revealing the density of lymphatics vessels may optimize the prognostic value of the International IgA predicting tool to calculate the risk of ESKD, favoring the evaluation of new targeted therapies.
Collapse
Affiliation(s)
- Lida Rodas
- Department of Nephrology and Renal Transplantation, Hospital Clínic, Department of Medicine, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
| | - Esther Barnadas
- Department of Pathology, Hospital Clínic, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
| | - Arturo Pereira
- Department of Hematology, Hospital Clínic, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Natalia Castrejon
- Department of Pathology, Hospital Clínic, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
| | - Anna Saurina
- Department of Nephrology, Hospital Terrasa, Cataluña, Spain
| | - Jordi Calls
- Department of Nephrology, Hospital de Mollet, Cataluña, Spain
| | - Yolanda Calzada
- Department of Nephrology, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Álvaro Madrid
- Department of Nephrology, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Miquel Blasco
- Department of Nephrology and Renal Transplantation, Hospital Clínic, Department of Medicine, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
| | - Esteban Poch
- Department of Nephrology and Renal Transplantation, Hospital Clínic, Department of Medicine, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
| | - Adriana García-Herrera
- Department of Pathology, Hospital Clínic, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
| | - Luis F. Quintana
- Department of Nephrology and Renal Transplantation, Hospital Clínic, Department of Medicine, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
| | - Catalán Group for the Study of Glomerular Diseases (GLOMCAT)
- Department of Nephrology and Renal Transplantation, Hospital Clínic, Department of Medicine, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
- Department of Pathology, Hospital Clínic, University of Barcelona, IDIBAPS and Centro de Referencia en Enfermedad Glomerular Compleja del Sistema Nacional de Salud de España (CSUR), Barcelona, Spain
- Department of Hematology, Hospital Clínic, University of Barcelona, IDIBAPS, Barcelona, Spain
- Department of Nephrology, Hospital Terrasa, Cataluña, Spain
- Department of Nephrology, Hospital de Mollet, Cataluña, Spain
- Department of Nephrology, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| |
Collapse
|
32
|
Lin F, Han S, Yu W, Rao T, Ruan Y, Yuan R, Li H, Ning J, Xia Y, Xie J, Qi Y, Zhou X, Cheng F. microRNA‐486‐5p is implicated in the cisplatin‐induced apoptosis and acute inflammation response of renal tubular epithelial cells by targeting HAT1. J Biochem Mol Toxicol 2022; 36:e23039. [PMID: 35279909 DOI: 10.1002/jbt.23039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 01/19/2022] [Accepted: 03/02/2022] [Indexed: 01/01/2023]
Affiliation(s)
- Fang‐You Lin
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Shang‐Ting Han
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Wei‐Min Yu
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Ting Rao
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Yuan Ruan
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Run Yuan
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Hao‐Yong Li
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Jin‐Zhuo Ning
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Yu‐Qi Xia
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Jin‐Na Xie
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Yu‐Cheng Qi
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Xiang‐Jun Zhou
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Fan Cheng
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| |
Collapse
|
33
|
Li J, Gong X. Tetramethylpyrazine: An Active Ingredient of Chinese Herbal Medicine With Therapeutic Potential in Acute Kidney Injury and Renal Fibrosis. Front Pharmacol 2022; 13:820071. [PMID: 35145414 PMCID: PMC8821904 DOI: 10.3389/fphar.2022.820071] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/10/2022] [Indexed: 12/23/2022] Open
Abstract
As an increasing public health concern worldwide, acute kidney injury (AKI) is characterized by rapid deterioration of kidney function. Although continuous renal replacement therapy (CRRT) could be used to treat severe AKI, effective drug treatment methods for AKI are largely lacking. Tetramethylpyrazine (TMP) is an active ingredient of Chinese herb Ligusticum wallichii (Chuan Xiong) with antioxidant and anti-inflammatory functions. In recent years, more and more clinical and experimental studies suggest that TMP might effectively prevent AKI. The present article reviews the potential mechanisms of TMP against AKI. Through search and review, a total of 23 studies were finally included. Our results indicate that the undergoing mechanisms of TMP preventing AKI are mainly related to reducing oxidative stress injury, inhibiting inflammation, preventing apoptosis of intrinsic renal cells, and regulating autophagy. Meanwhile, given that AKI and chronic kidney disease (CKD) are very tightly linked by each other, and AKI is also an important inducement of CKD, we thus summarized the potential of TMP impeding the progression of CKD through anti-renal fibrosis.
Collapse
|
34
|
Assessing and counteracting fibrosis is a cornerstone of the treatment of CKD secondary to systemic and renal limited autoimmune disorders. Autoimmun Rev 2021; 21:103014. [PMID: 34896651 DOI: 10.1016/j.autrev.2021.103014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is an increasing cause of morbidity and mortality worldwide. Besides the higher prevalence of diabetes, hypertension and aging worldwide, immune mediated disorders remain an important cause of kidney disease and are especially prevalent in young adults. Regardless of the initial insult, final pathway to CKD and kidney failure is always the loss of normal tissue and fibrosis development, in which the dynamic equilibrium between extracellular matrix synthesis and degradation is disturbed, leading to excessive production and accumulation. During fibrosis, a multitude of cell types intervene at different levels, but myofibroblasts and inflammatory cells are considered critical in the process. They exert their effects through different molecular pathways, of which transforming growth factor β (TGF-β) has demonstrated to be of particular importance. Additionally, CKD itself promotes fibrosis due to the accumulation of toxins and hormonal changes, and proteinuria is simultaneously a manifestation of CKD and a specific driver of renal fibrosis. Pathways involved in renal fibrosis and CKD are closely interrelated, and although important advances have been made in our knowledge of them, it is still necessary to translate them into clinical practice. Given the complexity of this process, it is highly likely that its treatment will require a multi-target strategy to control the origin of the damage but also the mechanisms that perpetuate it. Fortunately, rapid technology development over the last years and new available drugs in the nephrologist's armamentarium give reasons for optimism that more personalized assistance for CKD and renal fibrosis will appear in the future.
Collapse
|
35
|
Zhu YL, Huang J, Chen XY, Xie J, Yang Q, Wang JF, Deng XM. Senkyunolide I alleviates renal Ischemia-Reperfusion injury by inhibiting oxidative stress, endoplasmic reticulum stress and apoptosis. Int Immunopharmacol 2021; 102:108393. [PMID: 34857480 DOI: 10.1016/j.intimp.2021.108393] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/14/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ligusticum striatum DC. is traditionally used to treat ischemic diseases because of its potent effect against blood stasis and thrombosis, including various cardiovascular, cerebral and renal diseases. Senkyunolide I (SEI), which is the major active phthalide ingredient of Ligusticum striatum DC., is mainly distributed in kidney and has been shown to attenuate ischemia reperfusion injury in liver. However, the underlying effect of SEI against renal ischemia-reperfusion injury (IRI) remain unclear. METHODS Renal ischemia reperfusion mice model was established by clamping bilateral renal pedicles. In vitro oxidative stress model was induced by H2O2. Level of blood urea nitrogen (BUN) and serum creatinine (SCr) was tested for in vivo model evaluation, while cell viability was tested using CCK8 to evaluate in vitro model. SEI solution containing 1% DMSO was injected intraperitoneally in the I/R group, while normal saline containing 1% DMSO injected in the Sham group. Reduced glutathione (GSH) solution containing 1% DMSO was used as a positive control. RESULTS SEI protected renal function and structural integrity. It reversed the I/R-induced elevation of BUN, SCr levels and renal pathological injury. The secretion of proinflammatory cytokines including TNF-α and IL-6 was inhibited, and the renal apoptosis was attenuated by SEI. In addition, SEI played a protective role by reducing the production of reactive oxidative species (ROS), as shown by the elevated expression of antioxidant proteins including Nrf2, HO-1, NQO1, and reduced expression of endoplasmic reticulum stress (ERS) related proteins including GRP78 and CHOP. It also attenuated HK2 cell injury in an in vitro model induced by H2O2. CONCLUSIONS SEI alleviates renal injury induced by ischemia reperfusion with anti-inflammatory, anti-endoplasmic reticulum stress, anti-oxidative and anti-apoptotic effect.
Collapse
Affiliation(s)
- Ya-Lin Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Huang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xue-Ying Chen
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jian Xie
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qing Yang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jia-Feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Xiao-Ming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
36
|
Yang YY, Ye L, Chen J, Qiu Y, Yin YL, Li P. Dok3 is involved in cisplatin-induced acute kidney injury via regulation of inflammation and apoptosis. Biochem Biophys Res Commun 2021; 569:132-138. [PMID: 34245977 DOI: 10.1016/j.bbrc.2021.06.097] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/20/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022]
Abstract
Cisplatin-induced acute kidney injury (AKI) is associated with high morbidity and mortality worldwide, but the underlying mechanisms are not fully understood. Downstream-of-kinase 3 (Dok3), a member of the Dok family of adaptor proteins plays a critical role in inflammatory response and immune regulation; however, the role of Dok3 in cisplatin-induced AKI remains unclear. This study explored the effect and potential molecular mechanisms of Dok3 in cisplatin-induced AKI using Dok3 knockout (Dok3-/-) and control mice (129S) with or without administration of a single intraperitoneal injection of cisplatin. Apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, lactate dehydrogenase (LDH) release, and Hoechst staining. Inflammatory factors were measured using ELISA kits. Protein and gene expression levels were measured by western blot analysis and real-time PCR, respectively. The results showed that Dok3 was expressed in renal tubular epithelial cells. Dok3 expression was decreased in kidneys of mice treated with cisplatin and cisplatin-treated HK2 cells. Dok3-/- mice showed lower creatinine levels and NGAL expression, and increased survival rates compared to 129S mice. Cisplatin-induced production of TNF-α and IL-6, and renal tubular cell apoptosis was attenuated in Dok3-/- mice. In vitro experiments demonstrated that HK2 cells overexpressing Dok3 exhibited exacerbated cisplatin-induced apoptosis and production of TNF-α and IL-6. These findings demonstrate that Dok3 regulates cisplatin-induced AKI by regulating apoptosis and inflammation.
Collapse
Affiliation(s)
- Yong-Yu Yang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Center of Translational Medical and Innovative Drug, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| | - Ling Ye
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jing Chen
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yue Qiu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, 453003, China
| | - Ya-Ling Yin
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, 453003, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, 453003, China.
| |
Collapse
|
37
|
Wu R, Li J, Tu G, Su Y, Zhang X, Luo Z, Rong R, Zhang Y. Comprehensive Molecular and Cellular Characterization of Acute Kidney Injury Progression to Renal Fibrosis. Front Immunol 2021; 12:699192. [PMID: 34777334 PMCID: PMC8586649 DOI: 10.3389/fimmu.2021.699192] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/01/2021] [Indexed: 02/05/2023] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) represent different stages of renal failure; thus, CKD can be regarded as a result of AKI deterioration. Previous studies have demonstrated that immune cell infiltration, oxidative stress, and metabolic mentalism can support renal fibrosis progression in AKI cases. However, the most important triggers and cell types involved in this pathological progression remain unclear. This study was conducted to shed light into the underlying cellular and molecular features of renal fibrosis progression through the analysis of three mouse whole kidney and one human single-cell RNA-sequencing datasets publicly available. According to the different causes of AKI (ischemia reperfusion injury [IRI] or cisplatin), the mouse samples were divided into the CIU [control-IRI-unilateral ureteral obstruction (UUO)] and CCU (control-cisplatin-UUO) groups. Comparisons between groups revealed eight different modules of differentially expressed genes (DEGs). A total of 1,214 genes showed the same expression pattern in both CIU and CCU groups; however, 1,816 and 1,308 genes were expressed specifically in the CCU and CIU groups, respectively. Further assessment of the DEGs according to the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathway and Gene Ontology (GO) showed that T-cell activation, fatty acid metabolic process, and arachidonic acid metabolism were involved in the fibrosis progression in CIU and CCU. Single-cell RNA-sequencing data along with the collected DEGs information also revealed that the T-cell activation mainly happened in immune cells, whereas the fatty acid metabolic process and arachidonic acid metabolism occurred in tubule cells. Taken together, these findings suggest that the fibrosis process differed between the CIU and CCU stages, in which immune and tubule cells have different functions. These identified cellular and molecular features of the different stages of fibrosis progression may pave the way for exploring novel potential therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Renyan Wu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiawei Li
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Guowei Tu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Su
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuepeng Zhang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhe Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yi Zhang, ; Ruiming Rong, ; Zhe Luo,
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
- *Correspondence: Yi Zhang, ; Ruiming Rong, ; Zhe Luo,
| | - Yi Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
- Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yi Zhang, ; Ruiming Rong, ; Zhe Luo,
| |
Collapse
|