1
|
Zhang X, Shao W, Gao Y, Wang X. Macrophage polarization-mediated PKM2/mTORC1/YME1L signaling pathway activation in fibrosis associated with Cardiorenal syndrome. Cell Signal 2025; 131:111664. [PMID: 39961408 DOI: 10.1016/j.cellsig.2025.111664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Cardiorenal syndrome (CRS) is a complex condition characterized by the interplay between cardiac and renal dysfunction, often culminating in renal fibrosis. The role of macrophage polarization and its downstream effects in CRS-induced renal fibrosis remains an area of active investigation. METHODS Single-cell RNA sequencing (scRNA-seq) and immune infiltration analyses were employed to identify key immune cells and genes involved in renal fibrosis in CRS. Meta-analysis and pseudo-time analysis were conducted to validate the functional relevance of these genes. Functional studies utilizing CRISPR/Cas9 gene editing and lentiviral vectors assessed macrophage polarization and epithelial-to-mesenchymal transition (EMT). In vivo, a CRS mouse model was established, and fibrosis progression was tracked using histological and imaging methods. RESULTS The PKM2/mTORC1/YME1L signaling axis was identified as a critical pathway driving renal fibrosis, mediated by HIF-1α-induced M1 macrophage polarization. Inhibition of HIF-1α significantly alleviated renal fibrosis by restricting M1 polarization and suppressing the PKM2/mTORC1/YME1L axis. Co-culture models further demonstrated the involvement of EMT and metabolic reprogramming in affected cells. CONCLUSION Targeting the HIF-1α signaling pathway offers a promising therapeutic strategy for renal fibrosis by modulating macrophage polarization and the PKM2/mTORC1/YME1L axis.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China.
| | - Wen Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yun Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xiaojun Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
2
|
Burfeind KG, Funahashi Y, Su XT, Lackey AE, Hagen MW, Blanche S, Emathinger JM, Hebert JF, McDonough AA, Gurley SB, Nelson JW, Hutchens MP. Kidney cell response to acute cardiorenal and isolated kidney ischemia-reperfusion injury. Physiol Genomics 2025; 57:266-278. [PMID: 39982410 DOI: 10.1152/physiolgenomics.00161.2024] [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: 10/22/2024] [Revised: 11/29/2024] [Accepted: 02/14/2025] [Indexed: 02/22/2025] Open
Abstract
Acute cardiorenal syndrome (CRS) represents a critical intersection of cardiac and renal dysfunction with profound clinical implications. Despite its significance, the molecular underpinnings that mediate cellular responses within the kidney during CRS remain inadequately understood. We used single nucleus RNA sequencing (snRNAseq) to dissect the cellular transcriptomic landscape of the kidney following a translational model of CRS, cardiac arrest/cardiopulmonary resuscitation (CA/CPR) in comparison to ischemia-reperfusion injury (IRI). In each dataset, we found that proximal tubule (PT) cells of the kidney undergo significant gene expression changes, with decreased expression of genes critically important for cell identity and function, indicative of dedifferentiation. Based on this, we created a novel score to capture the dedifferentiation state of each kidney cell population and found that certain epithelial cell populations, such as the PT S1 and S2 segments, as well as the distal convoluted tubule, exhibited significant dedifferentiation response. Interestingly, the dedifferentiation response in the distal nephron differed in magnitude between IRI and CA/CPR. Gene set enrichment analysis (GSEA) of PT response to IRI and CA/CPR revealed similarities between the two models and key differences, including enrichment of immune system process genes. Transcriptional changes in both mouse models of acute kidney injury (AKI) highly correlated with a dataset of human biopsies from patients diagnosed with AKI. This comprehensive single-nucleus transcriptomic profiling provides valuable insights into the cellular mechanisms driving CRS.NEW & NOTEWORTHY Cardiac dysfunction is a common cause of acute kidney injury in a malady called acute cardiorenal syndrome. In a mouse model of acute cardiorenal syndrome called cardiac arrest/cardiopulmonary resuscitation, we characterized, for the first time, the kidney transcriptional landscape at the single-cell level. We developed a novel method for quantifying cell response to injury and found that cells adapted through dedifferentiation, the magnitude of which varied depending on cell type.
Collapse
Affiliation(s)
- Kevin G Burfeind
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Yoshio Funahashi
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Anne E Lackey
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Matt W Hagen
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
| | - Sienna Blanche
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Jacqueline M Emathinger
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Jessica F Hebert
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Susan B Gurley
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Jonathan W Nelson
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Michael P Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States
- Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, United States
| |
Collapse
|
3
|
Zhao X, Wang L. mTOR/p70S6K signaling pathway promotes fibrillin-1 expression in AKI-to-CKD transition post CA/CPR. Cell Signal 2025; 128:111624. [PMID: 39880104 DOI: 10.1016/j.cellsig.2025.111624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
The possible involvement of mTOR/p70S6K signaling in mediating Fibrillin-1 expression during the transition from acute kidney injury (AKI) to chronic kidney disease (CKD) after cardiac arrest and cardiopulmonary resuscitation (CA/CPR). A CA/CPR AKI model was established using male C57BL/6 mice aged 8-12 weeks. The expression of Fibrillin-1 and activation of the mTOR/p70S6K signaling pathway in kidney tissues were assessed at different time points. Rapamycin, administered intraperitoneally, inhibited the mTOR/p70S6K signaling pathway in CA/CPR AKI mice. Tissue immunofluorescence and immunohistochemistry were used to detect the injury, fibrosis, and inflammatory cell infiltration in renal tissues. The expression level of Fibrillin-1 and components of the mTOR/p70S6K signaling pathway, while ELISA quantified levels of inflammatory factors in renal tissues. Results showed that Fibrillin-1 expression progressively increased alongside enhanced mTOR/p70S6K signaling in the renal tissues of CA/CPR AKI mice. Inhibition of mTOR/p70S6K signaling by rapamycin reduced Fibrillin-1 expression, collagen deposition, and α-SMA levels, alleviating renal injury and decreasing macrophage and T cell infiltration, as well as inflammatory factor production. Conversely, combining rapamycin with Fibrillin-1 overexpression exacerbated renal injury and increased inflammatory factor production. Activation of the mTOR/p70S6K pathway upregulates Fibrillin-1 expression, potentially facilitating the progression from AKI to CKD in CA/CPR mice.
Collapse
Affiliation(s)
- Xiaohui Zhao
- School of Basic Medicine, Jiamusi University, Jiamusi 154007, PR China
| | - Limin Wang
- School of Basic Medicine, Jiamusi University, Jiamusi 154007, PR China.
| |
Collapse
|
4
|
Moronge D, Godley H, Ayulo V, Mellott E, Elgazzaz M, Cooper G, Mohamed R, Ogbi S, Gillis E, Faulkner JL, Sullivan JC. Persistent subclinical renal injury in female rats following renal ischemia-reperfusion injury. Clin Sci (Lond) 2025; 139:CS20241851. [PMID: 39902555 DOI: 10.1042/cs20241851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/05/2025]
Abstract
The incidence of acute kidney injury (AKI) continues to rise in both men and women. Although creatinine levels return to normal quicker in females following AKI than in males, it remains unclear whether subclinical renal injury persists in young females post-AKI. This study tested the hypothesis that AKI results in subclinical renal injury in females despite plasma creatinine returning to sham levels. For the present study, 12-13-week-old female Sprague-Dawley (SD) rats were randomized to sham or 45-minute warm bilateral ischemia-reperfusion surgery as an experimental model of ischemic AKI. Rats were euthanized 1, 3, 7, 14, or 30 days post-AKI/sham. Plasma creatinine, cystatin C, kidney injury molecule 1 (KIM-1), and NGAL were quantified via assay kits or immunoblotting. Kidneys were processed for histological analysis to assess tubular injury and fibrosis, and for electron microscopy to examine mitochondrial morphology. Immunoblots on kidney homogenates were performed to determine oxidative stress and apoptosis. Plasma creatinine levels were increased 24 hours post-AKI but returned to sham control levels three days post-AKI. However, cystatin C, KIM-1, and NGAL were increased 30 days post-AKI compared with sham. Tubular injury, tubulointerstitial fibrosis, and mitochondrial dysfunction were all increased in 30-day post-AKI rats compared with sham. Additionally, 30-day post-AKI rats had higher p-JNK expression and lower antioxidant enzyme glutathione peroxidase and catalase levels compared with sham. AKI resulted in higher expression of cleaved caspase 3, TUNEL+ cells, and caspase 9 than sham. Despite the normalization of creatinine levels, our data support the hypothesis that subclinical renal injury persists following ischemia-reperfusion injury in young female rats.
Collapse
Affiliation(s)
- Desmond Moronge
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Hannah Godley
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Victor Ayulo
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Elisabeth Mellott
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Mona Elgazzaz
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Gibson Cooper
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Riyaz Mohamed
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Safia Ogbi
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Ellen Gillis
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
- Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, U.S.A
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| |
Collapse
|
5
|
Wang X, Luo T, Yang Y, Yang L, Liu M, Zou Q, Wang D, Yang C, Xue Q, Liu S, Wan J, He G, Zeng A, Hou J, Ma S, Wang P. TRPA1 protects against contrast-induced renal tubular injury by preserving mitochondrial dynamics via the AMPK/DRP1 pathway. Free Radic Biol Med 2024; 224:521-539. [PMID: 39278575 DOI: 10.1016/j.freeradbiomed.2024.09.012] [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: 07/03/2024] [Revised: 08/22/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Mitochondrial dysfunction and oxidative stress are involved in the development of contrast-induced acute kidney injury (CI-AKI). The present study aimed to reveal the role of transient receptor potential ankyrin 1 (TRPA1), an oxidative sensor, in CI-AKI. Trpa1PT-/- mice with Trpa1 conditionally knocked out in renal proximal tubular (PT) cells, Trpa1 overexpression mice (Trpa1-OE), and TRPA1 agonists and antagonists were used to study its function in a mouse model of iohexol-induced CI-AKI. We found that TRPA1 was functionally expressed in PT cells. Activation of TRPA1 with cinnamaldehyde or overexpression of Trpa1 remarkably ameliorated renal tubular injury and dysfunction in a mouse model of CI-AKI, while CI-AKI was significantly exacerbated in Trpa1PT-/- mice. Proteomics demonstrated that mouse kidneys with CI-AKI had downregulated proteins involved in mitochondrial dynamics and upregulated mitophagy-associated proteins. The beneficial effects of TRPA1 activation/overexpression on CI-AKI were associated with improved mitochondrial function, decreased mitochondrial fission and oxidative stress, enhanced mitophagy, and less apoptosis of renal tubular cells. TRPA1-induced decreases in mitochondrial fission were linked to upregulated fusion-related proteins (mitofusin 1, mitofusin 2 and optic atrophy 1) and downregulated fission mediator, phosphorylated dynamin-related protein 1 (Drp1). Importantly, inhibition of Drp1 with mitochondrial division inhibitor 1 improved CI-AKI. In addition, the decreased mitochondrial fission was also mediated by inactivation of AMP-activated protein kinase which mediates mitochondrial biogenesis. The findings suggest that TRPA1 plays a protective role in CI-AKI through regulating mitochondrial fission/fusion, biogenesis, and dysfunction. Activating TRPA1 may become novel therapeutic strategies for the prevention of CI-AKI.
Collapse
Affiliation(s)
- Xinquan Wang
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Tao Luo
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Yi Yang
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Lun Yang
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Min Liu
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Qingliang Zou
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Dan Wang
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Changqiang Yang
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Qiang Xue
- Department of Cardiology, Yanan Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650051, China
| | - Sen Liu
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Jindong Wan
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Gaomin He
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Anping Zeng
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Jixin Hou
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China
| | - Shuangtao Ma
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| | - Peijian Wang
- Department of Cardiology, Department of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China; Key Laboratory of Aging and Vascular Homeostasis at Chengdu Medical College of Sichuan Province, Chengdu, Sichuan, 610500, China; Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, Sichuan, 610500, China.
| |
Collapse
|
6
|
Liu L, Liu Y, Xin Y, Liu Y, Gao Y, Yu K, Wang C. An early and stable mouse model of polymyxin-induced acute kidney injury. Intensive Care Med Exp 2024; 12:88. [PMID: 39352603 PMCID: PMC11445218 DOI: 10.1186/s40635-024-00667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Polymyxins have been revived as a last-line therapeutic option for multi-drug resistant bacteria and continue to account for a significant proportion of global antibiotic usage. However, kidney injury is often a treatment limiting event with kidney failure rates ranging from 5 to 13%. The mechanisms underlying polymyxin-induced nephrotoxicity are currently unclear. Researches of polymyxin-associated acute kidney injury (AKI) models need to be more standardized, which is crucial for obtaining consistent and robust mechanistic results. METHODS In this study, male C57BL/6 mice received different doses of polymyxin B (PB) and polymyxin E (PE, also known as colistin) by different routes once daily (QD), twice daily (BID), and thrice daily (TID) for 3 days. We continuously monitored the glomerular filtration rate (GFR) and the AKI biomarkers, including serum creatinine (Scr), blood urea nitrogen (BUN), neutrophil gelatinase-associated lipocalin (NGAL), and kidney injury molecule-1 (KIM-1). We also performed histopathological examinations to assess the extent of kidney injury. RESULTS Mice receiving PB (35 mg/kg/day subcutaneously) once daily exhibited a significant decrease in GFR and a notable increase in KIM-1 two hours after the first dose. Changes in GFR and KIM-1 at 24, 48 and 72 h were consistent and demonstrated the occurrence of kidney injury. Histopathological assessments showed a positive correlation between the severity of kidney injury and the changes in GFR and KIM-1 (Spearman's rho = 0.3167, P = 0.0264). The other groups of mice injected with PB and PE did not show significant changes in GFR and AKI biomarkers compared to the control group. CONCLUSION The group receiving PB (35 mg/kg/day subcutaneously) once daily consistently developed AKI at 2 h after the first dose. Establishing an early and stable AKI model facilitates researches into the mechanisms of early-stage kidney injury. In addition, our results indicated that PE had less toxicity than PB and mice receiving the same dose of PB in the QD group exhibited more severe kidney injury than the BID and TID groups.
Collapse
Affiliation(s)
- Linqiong Liu
- Departments of Critical Care Medicine, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
- Departments of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yuxi Liu
- Departments of Critical Care Medicine, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
- Departments of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yu Xin
- Departments of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yanqi Liu
- Departments of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yan Gao
- Departments of Critical Care Medicine, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| | - Kaijiang Yu
- Departments of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Changsong Wang
- Departments of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
7
|
Funahashi Y, Park SH, Hebert JF, Eiwaz MB, Munhall AC, Groat T, Zeng L, Kim J, Choi HS, Hutchens MP. Nanotherapeutic kidney cell-specific targeting to ameliorate acute kidney injury. Kidney Int 2024; 106:597-610. [PMID: 39067856 DOI: 10.1016/j.kint.2024.06.021] [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: 07/12/2023] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024]
Abstract
Acute kidney injury (AKI) increases the risk of in-hospital death, adds to expense of care, and risk of early chronic kidney disease. AKI often follows an acute event such that timely treatment could ameliorate AKI and potentially reduce the risk of additional disease. Despite therapeutic success of dexamethasone in animal models, clinical trials have not demonstrated broad success. To improve the safety and efficacy of dexamethasone for AKI, we developed and characterized a novel, kidney-specific nanoparticle enabling specific within-kidney targeting to proximal tubular epithelial cells provided by the megalin ligand cilastatin. Cilastatin and dexamethasone were complexed to H-Dot nanoparticles, which were constructed from generally recognized as safe components. Cilastatin/Dexamethasone/H-Dot nanotherapeutics were found to be stable at plasma pH and demonstrated salutary release kinetics at urine pH. In vivo, they were specifically biodistributed to the kidney and bladder, with 75% recovery in the urine and with reduced systemic toxicity compared to native dexamethasone. Cilastatin complexation conferred proximal tubular epithelial cell specificity within the kidney in vivo and enabled dexamethasone delivery to the proximal tubular epithelial cell nucleus in vitro. The Cilastatin/Dexamethasone/H-Dot nanotherapeutic improved kidney function and reduced kidney cellular injury when administered to male C57BL/6 mice in two translational models of AKI (rhabdomyolysis and bilateral ischemia reperfusion). Thus, our design-based targeting and therapeutic loading of a kidney-specific nanoparticle resulted in preservation of the efficacy of dexamethasone, combined with reduced off-target disposition and toxic effects. Hence, our study illustrates a potential strategy to target AKI and other diseases of the kidney.
Collapse
Affiliation(s)
- Yoshio Funahashi
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Seung Hun Park
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica F Hebert
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Mahaba B Eiwaz
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Adam C Munhall
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Tahnee Groat
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Lingxue Zeng
- Department of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Jonghan Kim
- Department of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael P Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA; Operative Care Division, Portland VA Medical Center, Portland, Oregon, USA.
| |
Collapse
|
8
|
Kundu S, Gairola S, Verma S, Mugale MN, Sahu BD. Chronic kidney disease activates the HDAC6-inflammatory axis in the heart and contributes to myocardial remodeling in mice: inhibition of HDAC6 alleviates chronic kidney disease-induced myocardial remodeling. Basic Res Cardiol 2024; 119:831-852. [PMID: 38771318 DOI: 10.1007/s00395-024-01056-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/22/2024]
Abstract
Chronic kidney disease (CKD) adversely affects the heart. The underlying mechanism and the interplay between the kidney and the heart are still obscure. We examined the cardiac effect using the unilateral ureteral obstruction (UUO)-induced CKD pre-clinical model in mice. Echocardiography, histopathology of the heart, myocardial mRNA expression of ANP and BNP, the extent of fibrotic (TGF-β, α-SMA, and collagen I) and epigenetic (histone deacetylases, namely HDAC3, HDAC4, and HDAC6) proteins, and myocardial inflammatory response were assessed. Six weeks of post-UUO surgery, we observed a compromised left-ventricular wall thickness and signs of cardiac hypertrophy, accumulation of fibrosis associated, and inflammatory proteins in the heart. In addition, we observed a perturbation of epigenetic proteins, especially HDAC3, HDAC4, and HDAC6, in the heart. Pharmacological inhibition of HDAC6 using ricolinostat (RIC) lessened cardiac damage and improved left-ventricular wall thickness. The RIC treatment substantially restored the serum cardiac injury markers, namely creatine kinase-MB and lactate dehydrogenase (LDH) activities, ANP and BNP mRNA expression, and heart histological changes. The extent of myocardial fibrotic proteins, phospho-NF-κB (p65), and pro-inflammatory cytokines (TNF-α, IL-18, and IL-1β) were significantly decreased in the RIC treatment group. Further findings revealed the CKD-induced infiltration of CD3, CD8a, CD11c, and F4/80 positive inflammatory cells in the heart. Treatment with RIC substantially reduced the myocardial infiltration of these inflammatory cells. From these findings, we believe that CKD-induced myocardial HDAC6 perturbation has a deteriorative effect on the heart, and inhibition of HDAC6 can be a promising approach to alleviate CKD-induced myocardial remodeling.
Collapse
Affiliation(s)
- Sourav Kundu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, Assam, 781101, India
| | - Shobhit Gairola
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, Assam, 781101, India
| | - Smriti Verma
- Department of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226 031, India
| | - Madhav Nilakanth Mugale
- Department of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226 031, India
| | - Bidya Dhar Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, Assam, 781101, India.
| |
Collapse
|
9
|
Xu Z, Tang J, Xin Chen, Jin Y, Zhang H, Liang R. Associations of C-reactive protein-albumin-lymphocyte (CALLY) index with cardiorenal syndrome: Insights from a population-based study. Heliyon 2024; 10:e37197. [PMID: 39296012 PMCID: PMC11408039 DOI: 10.1016/j.heliyon.2024.e37197] [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: 07/30/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Background Cardiorenal syndrome (CRS) is a complex condition characterized by the interplay of immune imbalance and inflammation. The C-reactive protein-Albumin-lymphocyte (CALLY) CALLY index serves as a new immune-nutritional scoring system, but its predictive value for CRS remains to be established. Methods In this study, we analyzed data from 27,978 participants in National Health and Nutrition Examination Survey (NHANES) from 1999 to 2010. The CALLY index was calculated as the ratio of albumin to lymphocyte, divided by C-reactive protein (CRP) multiplied by 104. CRS was defined by the coexistence of cardiovascular disease and chronic kidney disease (eGFR <60 mL/min/1.73 m2). Multivariate weighted logistic regression models were employed to determine the odds ratio and 95 % confidence interval for the association between the CALLY index and CRS. Receiver operating characteristic (ROC) curves and restricted cubic spline (RCS) curves were used to assess the predictive efficacy and nonlinear relationship, respectively. Results The prevalence of CRS in the study population was 1.22 %. Our findings revealed a significant inverse relationship between the CALLY index and CRS risk, with lower CALLY index values being associated with a higher likelihood of CRS (OR = 0.95, 95 % CI = 0.94-0.96, P < 0.001). Participants in higher quartiles of the CALLY index showed a progressively reduced risk of CRS (P for trend <0.001). Moreover, the CALLY index demonstrated superior predictive performance compared to other inflammatory indicators, such as systemic immune-inflammation index (SII), neutrophil/high-density lipoprotein ratio (NHR), lymphocyte/high-density lipoprotein ratio (LHR), monocyte/high-density lipoprotein ratio (MHR), and platelet/high-density lipoprotein ratio (PHR) (AUC = 0.672, 95 % CI = 0.643-0.701). Conclusions This study underscores the significant negative correlation between the CALLY index and the risk of cardiorenal syndrome. The CALLY index emerges as a robust and independent predictor of CRS, outperforming traditional inflammatory markers. This finding highlights the potential utility of the CALLY index as a clinical tool for identifying individuals at risk for CRS.
Collapse
Affiliation(s)
- Zhehao Xu
- Department of General Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China
| | - Jiao Tang
- Department of Cardiovascular Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, China
| | - Xin Chen
- Department of Cardiovascular Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, China
| | - Yian Jin
- Department of General Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China
| | - Huanji Zhang
- Department of Cardiovascular Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, China
| | - Ruiyun Liang
- Department of Respiratory Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China
| |
Collapse
|
10
|
Puri B, Majumder S, Gaikwad AB. Significance of LncRNAs in AKI-to-CKD transition: A therapeutic and diagnostic viewpoint. Life Sci 2024; 342:122509. [PMID: 38387702 DOI: 10.1016/j.lfs.2024.122509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Acute kidney injury to chronic kidney disease (AKI-to-CKD) transition is a complex intermingling of characteristics of both AKI and CKD. Pathophysiologically, the transition lasts seven days after the AKI episode and thereafter silently progresses towards CKD. Growing reports confirm that the AKI-to-CKD transition is heavily regulated by epigenetic modifiers. Long non-coding RNAs (lncRNAs) share a diverse role in gene regulation at transcriptional and translational levels and have been reported to be involved in the regulation and progression of AKI-to-CKD transition. Several lncRNAs have been considered potential biomarkers for diagnosing kidney disease, including AKI and CKD. Targeting lncRNAs gives a promising therapeutic strategy against kidney diseases. The primitive role of lncRNA in the progression of the AKI-to-CKD transition is yet to be fully understood. As known, the lncRNAs could be used as a biomarker and a therapeutic target to halt the CKD development and progression after AKI. This review aims to deepen our understanding of the current knowledge regarding the involvement of lncRNAs in the AKI-to-CKD transition. This review primarily discusses the role of lncRNAs and the change in their mechanisms during different stages of kidney disease, such as in AKI, AKI-to-CKD transition, and CKD. Further, we have discussed the potential diagnostic and pharmacological outcomes of targeting lncRNAs to prevent or slow the progression of AKI-to-CKD transition.
Collapse
Affiliation(s)
- Bhupendra Puri
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Syamantak Majumder
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
11
|
Burfeind KG, Funahashi Y, Munhall AC, Eiwaz M, Hutchens MP. Natural Killer Lymphocytes Mediate Renal Fibrosis Due to Acute Cardiorenal Syndrome. KIDNEY360 2024; 5:8-21. [PMID: 38037228 PMCID: PMC10833608 DOI: 10.34067/kid.0000000000000305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Key Points Natural killer cells infiltrate the kidney after cardiac arrest and medial renal fibrosis Granzyme A is produced by natural killer cells and causes mesenchymal cell expansion and fibrosis in type 1 cardiorenal syndrome Background The AKI to CKD transition presents an opportunity for intervention to prevent CKD. Our laboratory developed a novel murine model of AKI-CKD transition and cardiac arrest/cardiopulmonary resuscitation (CA/CPR), in which all animals develop CKD at 7 weeks. The purpose of this study was to identify potential immune drivers of fibrosis after CA/CPR. Methods Cardiac arrest was induced by potassium chloride, and mice were resuscitated with chest compressions and epinephrine. The kidney immune landscape after CA/CPR was profiled using 11-color flow cytometry analysis and immunofluorescence. Immune cell-derived mediators of fibrosis were identified by analyzing data from three previously published single-cell or single-nuclear RNA sequencing studies. NRK49F fibroblasts were treated with granzyme A (GzA) in vitro , and then cell proliferation was quantified using 5-ethynyl-2′-deoxyuridine. GzA was pharmacologically inhibited both in vitro and in vivo . Results Immune cells infiltrated the kidney after CA/CPR, consisting primarily of innate immune cells, including monocytes/macrophages, neutrophils, and natural killer (NK) cells. NK cell infiltration immediately preceded mesenchymal cell expansion, which occurred starting 7 days after CA/CPR. Immune cells colocalized with mesenchymal cells, accumulating in the areas of fibrosis. Analysis of previously published single-cell or single-nuclear RNA sequencing data revealed GzA as a potential mediator of immune to mesenchymal communication. GzA administration to fibroblasts in vitro induced cell growth and proliferation. Pharmacologic blockade of GzA signaling in vivo attenuated fibrosis and improved renal function after CA/CPR. Conclusions Renal inflammation occurs during cardiorenal syndrome, which correlates with mesenchymal cell expansion. GzA, produced by NK cells, presents a novel therapeutic target to prevent the transition to CKD after AKI.
Collapse
Affiliation(s)
- Kevin G. Burfeind
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
| | | | | | - Mahaba Eiwaz
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
- Portland VA Medical Center, Portland, Oregon
| | - Michael P. Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
- Portland VA Medical Center, Portland, Oregon
| |
Collapse
|
12
|
Gao L, Zhang J, Yang T, Jiang L, Liu X, Wang S, Wang X, Huang Y, Wang H, Zhang M, Gong T, Ma L, Li C, He C, Meng XM, Wu Y. STING/ACSL4 axis-dependent ferroptosis and inflammation promote hypertension-associated chronic kidney disease. Mol Ther 2023; 31:3084-3103. [PMID: 37533255 PMCID: PMC10556226 DOI: 10.1016/j.ymthe.2023.07.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023] Open
Abstract
Hypertension is a primary modifiable risk factor for cardiovascular diseases, which often induces renal end-organ damage and complicates chronic kidney disease (CKD). In the present study, histological analysis of human kidney samples revealed that hypertension induced mtDNA leakage and promoted the expression of stimulator of interferon genes (STING) in renal epithelial cells. We used angiotensin II (AngII)- and 2K1C-treated mouse kidneys to elucidate the underlying mechanisms. Abnormal renal mtDNA packing caused by AngII promoted STING-dependent production of inflammatory cytokines, macrophage infiltration, and a fibrogenic response. STING knockout significantly decreased nuclear factor-κB activation and immune cell infiltration, attenuating tubule atrophy and extracellular matrix accumulation in vivo and in vitro. These effects delayed CKD progression. Immunoprecipitation assays and liquid chromatography-tandem mass spectrometry showed that STING and ACSL4 were directly combined at the D53 and K412 amino acids of ACSL4. Furthermore, STING induced renal inflammatory response and fibrosis through ACSL4-dependent ferroptosis. Last, inhibition of ACSL4 using small interfering RNA, rosiglitazone, or Fer-1 downregulated AngII-induced mtDNA-STING-dependent renal inflammation. These results suggest that targeting the STING/ACSL4 axis might represent a potential strategy for treating hypertension-associated CKD.
Collapse
Affiliation(s)
- Li Gao
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Center for Scientific Research of Anhui Medical University, Hefei 230022, China
| | - Junsheng Zhang
- The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Anhui Public Health Clinical Center, Hefei 230032, China
| | - Tingting Yang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Jiang
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xueqi Liu
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Sheng Wang
- Center for Scientific Research of Anhui Medical University, Hefei 230022, China
| | - Xian Wang
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yuebo Huang
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Huaying Wang
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Mengya Zhang
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Tingting Gong
- The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Lijuan Ma
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Chao Li
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Chaoyong He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiao-Ming Meng
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Yonggui Wu
- Department of Nephropathy, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Center for Scientific Research of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
13
|
Chen J, Tang TT, Cao JY, Li ZL, Zhong X, Wen Y, Shen AR, Liu BC, Lv LL. KIM-1 augments hypoxia-induced tubulointerstitial inflammation through uptake of small extracellular vesicles by tubular epithelial cells. Mol Ther 2023; 31:1437-1450. [PMID: 35982620 PMCID: PMC10188645 DOI: 10.1016/j.ymthe.2022.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/29/2022] [Accepted: 08/12/2022] [Indexed: 12/19/2022] Open
Abstract
Tubular epithelial cells (TECs) exposed to hypoxia incite tubulointerstitial inflammation (TII), while the exact mechanism is unclear. In this study, we identified that hypoxia evoked tubule injury as evidenced by tubular hypoxia-inducible factor-1α and kidney injury molecule-1 (KIM-1) expression and that renal small extracellular vesicle (sEV) production was increased with the development of TII after ischemia-reperfusion injury (IRI). Intriguingly, KIM-1-positive tubules were surrounded by macrophages and co-localized with sEVs. In vitro, KIM-1 expression and sEV release were increased in hypoxic TECs and the hypoxia-induced inflammatory response was ameliorated when KIM-1 or Rab27a, a master regulator of sEV secretion, was silenced. Furthermore, KIM-1 was identified to mediate hypoxic TEC-derived sEV (Hypo-sEV) uptake by TECs. Phosphatidylserine (PS), a ligand of KIM-1, was present in Hypo-sEVs as detected by nanoflow cytometry. Correspondingly, the inflammatory response induced by exogenous Hypo-sEVs was attenuated when KIM-1 was knocked down. In vivo, exogenous-applied Hypo-sEVs localized to KIM-1-positive tubules and exacerbated TII in IRI mice. Our study demonstrated that KIM-1 expressed by injured tubules mediated sEV uptake via recognizing PS, which participated in the amplification of tubule inflammation induced by hypoxia, leading to the development of TII in ischemic acute kidney injury.
Collapse
Affiliation(s)
- Jun Chen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Jing-Yuan Cao
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Xin Zhong
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - An-Ran Shen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China.
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, 87 Ding Jia Qiao Road, Nanjing 210009, China.
| |
Collapse
|
14
|
Hebert JF, Funahashi Y, Hutchens MP. Harm! foul! How acute kidney injury SHReDDs patient futures. Curr Opin Nephrol Hypertens 2023; 32:165-171. [PMID: 36683541 PMCID: PMC10079264 DOI: 10.1097/mnh.0000000000000864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW Transition from acute kidney injury (AKI) to chronic kidney disease (CKD) is increasingly accepted. Less well recognized, but supported by very similar data, is development of disease of other organ systems after AKI. Awareness of other-organ sequelae of AKI may inform efforts to improve the care of patients after AKI. RECENT FINDINGS Stroke, hypertension, reproductive risk, dementia, and death (SHReDD) are sequelae, which occur with increased risk relative to that of non-AKI within 6 months-3 years after AKI diagnosis, and which are supported by preclinical/mechanistic study. Adjusted hazard ratios for these sequelae are strikingly similar to that of AKI-CKD, ranging from 1.2 to 3.0. Mechanistic studies suggest kidney-centric mechanisms including sodium regulation, volume status regulation, and the renin-angiotensin system are drivers of long-term, extra-renal, change. SUMMARY Further clinical characterization and mechanistic insight is necessary, and may have considerable translational impact. Programs which screen or follow post-AKI patients may increase clinical utility if focus is expanded to include the SHReDD complications.
Collapse
Affiliation(s)
- Jessica F Hebert
- Department of Anesthesiology & Perioperative Medicine, Oregon Health and Science University
| | - Yoshio Funahashi
- Department of Anesthesiology & Perioperative Medicine, Oregon Health and Science University
| | - Michael P Hutchens
- Department of Anesthesiology & Perioperative Medicine, Oregon Health and Science University
- Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, USA
| |
Collapse
|
15
|
Czopek A, Moorhouse R, Gallacher PJ, Pugh D, Ivy JR, Farrah TE, Godden E, Hunter RW, Webb DJ, Tharaux PL, Kluth DC, Dear JW, Bailey MA, Dhaun N. Endothelin blockade prevents the long-term cardiovascular and renal sequelae of acute kidney injury in mice. Sci Transl Med 2022; 14:eabf5074. [PMID: 36516266 DOI: 10.1126/scitranslmed.abf5074] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) is common and associated with increased risks of cardiovascular and chronic kidney disease. Causative molecular/physiological pathways are poorly defined. There are no therapies to improve long-term outcomes. An activated endothelin system promotes cardiovascular and kidney disease progression. We hypothesized a causal role for this in the transition of AKI to chronic disease. Plasma endothelin-1 was threefold higher; urine endothelin-1 was twofold higher; and kidney preproendothelin-1, endothelin-A, and endothelin-B receptor message up-regulated in patients with AKI. To show causality, AKI was induced in mice by prolonged ischemia with a 4-week follow-up. Ischemic injury resulted in hypertension, endothelium-dependent and endothelium-independent macrovascular and microvascular dysfunction, and an increase in circulating inflammatory Ly6Chigh monocytes. In the kidney, we observed fibrosis, microvascular rarefaction, and inflammation. Administration of endothelin-A antagonist, but not dual endothelin-A/B antagonist, normalized blood pressure, improved macrovascular and microvascular function, and prevented the transition of AKI to CKD. Endothelin-A blockade reduced circulating and renal proinflammatory Ly6Chigh monocytes and B cells, and promoted recruitment of anti-inflammatory Ly6Clow monocytes to the kidney. Blood pressure reduction alone provided no benefits; blood pressure reduction alongside blockade of the endothelin system was as effective as endothelin-A antagonism in mitigating the long-term sequelae of AKI in mice. Our studies suggest up-regulation of the endothelin system in patients with AKI and show in mice that existing drugs that block the endothelin system, particularly those coupling vascular support and anti-inflammatory action, can prevent the transition of AKI to chronic kidney and cardiovascular disease.
Collapse
Affiliation(s)
- Alicja Czopek
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Rebecca Moorhouse
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Peter J Gallacher
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Dan Pugh
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK
| | - Jessica R Ivy
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Tariq E Farrah
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK
| | - Emily Godden
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Robert W Hunter
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK
| | - David J Webb
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Centre (PARCC), Institut National de la Santé et de la Recherche Médicale (INSERM), 75015 Paris, France
| | - David C Kluth
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - James W Dear
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Matthew A Bailey
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Neeraj Dhaun
- Edinburgh Kidney, University/BHF Centre of Research Excellence, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.,Department of Renal Medicine, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK.,Paris Cardiovascular Research Centre (PARCC), Institut National de la Santé et de la Recherche Médicale (INSERM), 75015 Paris, France
| |
Collapse
|
16
|
Experimental models of acute kidney injury for translational research. Nat Rev Nephrol 2022; 18:277-293. [PMID: 35173348 DOI: 10.1038/s41581-022-00539-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
Abstract
Preclinical models of human disease provide powerful tools for therapeutic discovery but have limitations. This problem is especially apparent in the field of acute kidney injury (AKI), in which clinical trial failures have been attributed to inaccurate modelling performed largely in rodents. Multidisciplinary efforts such as the Kidney Precision Medicine Project are now starting to identify molecular subtypes of human AKI. In addition, over the past decade, there have been developments in human pluripotent stem cell-derived kidney organoids as well as zebrafish, rodent and large animal models of AKI. These organoid and AKI models are being deployed at different stages of preclinical therapeutic development. However, the traditionally siloed, preclinical investigator-driven approaches that have been used to evaluate AKI therapeutics to date rarely account for the limitations of the model systems used and have given rise to false expectations of clinical efficacy in patients with different AKI pathophysiologies. To address this problem, there is a need to develop more flexible and integrated approaches, involving teams of investigators with expertise in a range of different model systems, working closely with clinical investigators, to develop robust preclinical evidence to support more focused interventions in patients with AKI.
Collapse
|
17
|
Funahashi Y, Ikeda M, Wakasaki R, Chowdhury S, Groat T, Zeppenfeld D, Hutchens MP. Renal injury in cardiorenal syndrome type 1 is mediated by albumin. Physiol Rep 2022; 10:e15173. [PMID: 35150207 PMCID: PMC8838648 DOI: 10.14814/phy2.15173] [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: 10/25/2021] [Revised: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 11/24/2022] Open
Abstract
Cardiorenal syndrome type 1 (CRS‐1) acute kidney injury (AKI) is a critical complication of acute cardiovascular disease but is poorly understood. AKI induces acute albuminuria. As chronic albuminuria is associated with worsening kidney disease and albumin has been implicated in tubular epithelial injury, we investigated whether albumin participates in CRS‐1, and whether CRS‐1 alters renal albumin handling. We report the role of albumin in in vivo and in vitro CRS‐1 models. An established translational model, cardiac arrest and cardiopulmonary resuscitation (CA/CPR) induced severe acute albuminuria which correlated with tubular epithelial cell death. In vivo microscopy demonstrated CA/CPR‐induced glomerular filtration of exogenous albumin, while administration of exogenous albumin after CA/CPR worsened AKI compared to iso‐oncotic control. Increased albumin signal was observed in the proximal tubules of CA/CPR mice compared to sham. Comparison of albumin flux from tubular lumen to epithelial cells revealed saturated albumin transport within minutes of albumin injection after CA/CPR. In vitro, HK2 cells (human kidney tubular epithelial cells), exposed to oxygen‐glucose deprivation were injured by albumin in a dose dependent fashion. This interference was unchanged by the tubular endocytic receptor megalin. In conclusion, CRS‐1 alters albumin filtration and tubular uptake, leading to increased tubular exposure to albumin, which is injurious to tubular epithelial cells, worsening AKI. Our findings shed light on the pathophysiology of renal albumin and may guide interventions such as albumin resuscitation to improve CRS‐1 outcomes. This investigation may have important translational relevance for patients that receive exogenous albumin as part of their CRS‐1 treatment regimen.
Collapse
Affiliation(s)
- Yoshio Funahashi
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Mizuko Ikeda
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Rumie Wakasaki
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Sheuli Chowdhury
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Tahnee Groat
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Douglas Zeppenfeld
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael P Hutchens
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA.,Operative Care Division, Portland Veterans Affairs Medical Center, Portland, Oregon, USA
| |
Collapse
|
18
|
Huang J, Xu C. LncRNA MALAT1-deficiency restrains lipopolysaccharide (LPS)-induced pyroptotic cell death and inflammation in HK-2 cells by releasing microRNA-135b-5p. Ren Fail 2021; 43:1288-1297. [PMID: 34503385 PMCID: PMC8439250 DOI: 10.1080/0886022x.2021.1974037] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 02/09/2023] Open
Abstract
Long non-coding RNAs (LncRNAs) participate in the regulation of chronic kidney disease (CKD), and acute kidney injury (AKI) is identified as an important risk factor for CKD. This study investigated the involvement of a novel LncRNA MALAT1 in regulating lipopolysaccharide (LPS)-induced cell pyroptosis and inflammation in the human renal tubular epithelial HK-2 cells. Here, the HK-2 cells were subjected to LPS (2 μg/mL) treatment to establish cellular AKI models in vitro, and we validated that LPS triggered NLRP3-mediated pyroptotic cell death, promoted cell apoptosis and inflammation-associated cytokines secretion to induce HK-2 cell injury. Then, a novel LncRNA MALAT1/miRNA (miRNA)-135b-5p axis was verified to rescue cell viability in LPS treated HK-2 cells by targeting NLRP3. Mechanistically, miRNA-135b-5p bound to LncRNA MALAT1, and LncRNA MALAT1 positively regulated NLRP3 through acting as RNA sponger for miRNA-135b-5p. Further gain- and loss-of-function experiments evidenced that both LncRNA MALAT1 ablation and miRNA-135b-5p overexpression reversed LPS-induced cell pyroptosis, apoptosis, and inflammation in the HK-2 cells, and the protective effects of LncRNA MALAT1 knock-down on LPS-treated HK-2 cells were abrogated by silencing miRNA-135b-5p. In general, our study firstly investigated the role of the LncRNA MALAT1/ miRNA-135b-5p/NLRP3 signaling cascade in regulating LPS-induced inflammatory death in HK-2 cells.
Collapse
Affiliation(s)
- Jie Huang
- Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, PR China
| | - Chen Xu
- Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, PR China
| |
Collapse
|
19
|
Matsushita K, Mori K, Saritas T, Eiwaz MB, Funahashi Y, Nickerson MN, Hebert JF, Munhall AC, McCormick JA, Yanagita M, Hutchens MP. Cilastatin Ameliorates Rhabdomyolysis-induced AKI in Mice. J Am Soc Nephrol 2021; 32:2579-2594. [PMID: 34341182 PMCID: PMC8722809 DOI: 10.1681/asn.2020030263] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/17/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Rhabdomyolysis, the destruction of skeletal muscle, is a significant cause of AKI and death in the context of natural disaster and armed conflict. Rhabdomyolysis may also initiate CKD. Development of specific pharmacologic therapy is desirable because supportive care is nearly impossible in austere environments. Myoglobin, the principal cause of rhabdomyolysis-related AKI, undergoes megalin-mediated endocytosis in proximal tubule cells, a process that specifically injures these cells. METHODS To investigate whether megalin is protective in a mouse model of rhabdomyolysis-induced AKI, we used male C57BL/6 mice and mice (14-32 weeks old) with proximal tubule-specific deletion of megalin. We used a well-characterized rhabdomyolysis model, injection of 50% glycerol in normal saline preceded by water deprivation. RESULTS Inducible proximal tubule-specific deletion of megalin was highly protective in this mouse model of rhabdomyolysis-induced AKI. The megalin knockout mice demonstrated preserved GFR, reduced proximal tubule injury (as indicated by kidney injury molecule-1), and reduced renal apoptosis 24 hours after injury. These effects were accompanied by increased urinary myoglobin clearance. Unlike littermate controls, the megalin-deficient mice also did not develop progressive GFR decline and persistent new proteinuria. Administration of the pharmacologic megalin inhibitor cilastatin to wild-type mice recapitulated the renoprotective effects of megalin deletion. This cilastatin-mediated renoprotective effect was dependent on megalin. Cilastatin administration caused selective proteinuria and inhibition of tubular myoglobin uptake similar to that caused by megalin deletion. CONCLUSIONS We conclude that megalin plays a critical role in rhabdomyolysis-induced AKI, and megalin interference and inhibition ameliorate rhabdomyolysis-induced AKI. Further investigation of megalin inhibition may inform translational investigation of a novel potential therapy.
Collapse
Affiliation(s)
- Katsuyuki Matsushita
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Kiyoshi Mori
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Turgay Saritas
- Division of Nephrology & Hypertension, Oregon Health & Science University, Portland, Oregon,Division of Nephrology & Hypertension, University Hospital RWTH Aachen, Aachen, Germany
| | - Mahaba B. Eiwaz
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Yoshio Funahashi
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Megan N. Nickerson
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Jessica F. Hebert
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Adam C. Munhall
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - James A. McCormick
- Division of Nephrology & Hypertension, Oregon Health & Science University, Portland, Oregon
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan,Institute for Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Michael P. Hutchens
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon,Portland Veterans Affairs Medical Center, Operative Care Division, Portland, Oregon,Correspondence: Dr. Michael P. Hutchens, Operative Care Division, Portland Veterans Affairs Medical Center R&D, 5 3710 SW US Veterans Hospital Road, Portland, OR 97239.
| |
Collapse
|
20
|
Li L, Lee J, Cho A, Kim JH, Ju W, An JN, Park JH, Zhu SM, Lee J, Yu SS, Lim CS, Kim DK, Kim YS, Yang SH, Lee JP. cMet agonistic antibody prevents acute kidney injury to chronic kidney disease transition by suppressing Smurf1 and activating Smad7. Clin Sci (Lond) 2021; 135:1427-1444. [PMID: 34061176 DOI: 10.1042/cs20210013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 11/17/2022]
Abstract
We aimed to investigate the role of cMet agonistic antibody (cMet Ab) in preventing kidney fibrosis during acute kidney injury (AKI) to chronic kidney disease (CKD) transition. Additionally, we explored the effect of cMet Ab on TGF-β1/Smad pathway during the pathogenesis of kidney fibrosis. A unilateral ischemia-reperfusion injury (UIRI) mouse model was established to induce AKI-to-CKD transition. Furthermore, we incubated human proximal tubular epithelial cells (hPTECs) under hypoxic conditions as in vitro model of kidney fibrosis. We analyzed the soluble plasma cMet level in patients with AKI requiring dialysis. Patients who did not recover kidney function and progressed to CKD presented a higher increase in the cMet level. The kidneys of mice treated with cMet Ab showed fewer contractions and weighed more than the controls. The mice in the cMet Ab-treated group showed reduced fibrosis and significantly decreased expression of fibronectin and α-smooth muscle actin. cMet Ab treatment decreased inflammatory markers (MCP-1, TNF-α, and IL-1β) expression, reduced Smurf1 and Smad2/3 level, and increased Smad7 expressions. cMet Ab treatment increased cMet expression and reduced the hypoxia-induced increase in collagen-1 and ICAM-1 expression, thereby reducing apoptosis in the in vitro cell model. After cMet Ab treatment, hypoxia-induced expression of Smurf1, Smad2/3, and TGF-β1 was reduced, and suppressed Smad7 was activated. Down-regulation of Smurf1 resulted in suppression of hypoxia-induced fibronectin expression, whereas treatment with cMet Ab showed synergistic effects. cMet Ab can successfully prevent fibrosis response in UIRI models of kidney fibrosis by decreasing inflammatory response and inhibiting the TGF-β1/Smad pathway.
Collapse
Affiliation(s)
- Lilin Li
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Intensive Care Unit, Yanbian University Hospital, Yanji, Jilin, China
| | - Jeonghwan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Ara Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Jin Hyuk Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Wonmin Ju
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jung Nam An
- Department of Internal Medicine, Hallym Sacred Heart Hospital, Anyang, Gyeonggi-do, Republic of Korea
| | - Jeong Hwan Park
- Department of Pathology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Shi Mao Zhu
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Junghun Lee
- R&D Center of Innovative Medicines, Helixmith Co., Ltd., Seoul, Republic of Korea
| | - Seung-Shin Yu
- R&D Center of Innovative Medicines, Helixmith Co., Ltd., Seoul, Republic of Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Hee Yang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Seoul National University Kidney Research Institute, Seoul, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
- Seoul National University Kidney Research Institute, Seoul, Republic of Korea
| |
Collapse
|
21
|
Belliere J, Casemayou A, Colliou E, El Hachem H, Kounde C, Piedrafita A, Feuillet G, Schanstra JP, Faguer S. Ibrutinib does not prevent kidney fibrosis following acute and chronic injury. Sci Rep 2021; 11:11985. [PMID: 34099830 PMCID: PMC8184891 DOI: 10.1038/s41598-021-91491-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/25/2021] [Indexed: 01/28/2023] Open
Abstract
Recent studies suggested that ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor, developed for the treatment of chronic lymphocytic leukemia, may prevent NLRP3 inflammasome activation in macrophages, IL-1β secretion and subsequent development of inflammation and organ fibrosis. The role of NLRP3 has been underlined in the various causes of acute kidney injury (AKI), a pathology characterized by high morbimortality and risk of transition toward chronic kidney disease (CKD). We therefore hypothesized that the BTK-inhibitor ibrutinib could be a candidate drug for AKI treatment. Here, we observed in both an AKI model (glycerol-induced rhabdomyolysis) and a model of rapidly progressive kidney fibrosis (unilateral ureteral obstruction), that ibrutinib did not prevent inflammatory cell recruitment in the kidney and fibrosis. Moreover, ibrutinib pre-exposure led to high mortality rate owing to severer rhabdomyolysis and AKI. In vitro, ibrutinib potentiated or had no effect on the secretion of IL-1β by monocytes exposed to uromodulin or myoglobin, two danger-associated molecule patterns proteins involved in the AKI to CKD transition. According to these results, ibrutinib should not be considered a candidate drug for patients developing AKI.
Collapse
Affiliation(s)
- Julie Belliere
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Audrey Casemayou
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Eloïse Colliou
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Hélène El Hachem
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Clément Kounde
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Alexis Piedrafita
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France
| | - Guylène Feuillet
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
| | - Joost P Schanstra
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier-Toulouse 3, Toulouse, France
| | - Stanislas Faguer
- UMR 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires, Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France.
- Université Paul Sabatier-Toulouse 3, Toulouse, France.
- Département de Néphrologie Et Transplantation D'organes, Centre de Référence Des Maladies Rénales Rares, INSERM U1048 (I2MC, équipe 12), Centre Hospitalier Universitaire de Toulouse, 1, avenue du Pr Jean Poulhes, 31059, Toulouse, France.
| |
Collapse
|
22
|
Zhang Y, Jiang Y, Yang W, Shen L, He B. Chronic Secondary Cardiorenal Syndrome: The Sixth Innovative Subtype. Front Cardiovasc Med 2021; 8:639959. [PMID: 33768118 PMCID: PMC7985164 DOI: 10.3389/fcvm.2021.639959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/27/2021] [Indexed: 12/02/2022] Open
Affiliation(s)
- Yipeng Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wentao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Abstract
Interstitial fibrosis with tubule atrophy (IF/TA) is the response to virtually any sustained kidney injury and correlates inversely with kidney function and allograft survival. IF/TA is driven by various pathways that include hypoxia, renin-angiotensin-aldosterone system, transforming growth factor (TGF)-β signaling, cellular rejection, inflammation and others. In this review we will focus on key pathways in the progress of renal fibrosis, diagnosis and therapy of allograft fibrosis. This review discusses the role and origin of myofibroblasts as matrix producing cells and therapeutic targets in renal fibrosis with a particular focus on renal allografts. We summarize current trends to use multi-omic approaches to identify new biomarkers for IF/TA detection and to predict allograft survival. Furthermore, we review current imaging strategies that might help to identify and follow-up IF/TA complementary or as alternative to invasive biopsies. We further discuss current clinical trials and therapeutic strategies to treat kidney fibrosis.Supplemental Visual Abstract; http://links.lww.com/TP/C141.
Collapse
|
24
|
Huang C, Chen Y, Lai B, Chen YX, Xu CY, Liu YF. Overexpression of SP1 restores autophagy to alleviate acute renal injury induced by ischemia-reperfusion through the miR-205/PTEN/Akt pathway. JOURNAL OF INFLAMMATION-LONDON 2021; 18:7. [PMID: 33546692 PMCID: PMC7863508 DOI: 10.1186/s12950-021-00270-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/21/2021] [Indexed: 12/30/2022]
Abstract
Background Acute kidney injury (AKI) is a major kidney disease with poor clinical outcome. SP1, a well-known transcription factor, plays a critical role in AKI and subsequent kidney repair through the regulation of various cell biologic processes. However, the underlying mechanism of SP1 in these pathological processes remain largely unknown. Methods An in vitro HK-2 cells with anoxia-reoxygenation injury model (In vitro simulated ischemic injury disease) and an in vivo rat renal ischemia-reperfusion injury model were used in this study. The expression levels of SP1, miR-205 and PTEN were detected by RT-qPCR, and the protein expression levels of SP1, p62, PTEN, AKT, p-AKT, LC3II, LC3I and Beclin-1 were assayed by western blot. Cell proliferation was assessed by MTT assay, and the cell apoptosis was detected by flow cytometry. The secretions of IL-6 and TNF-α were detected by ELISA. The targeted relationship between miR-205 and PTEN was confirmed by dual luciferase report assay. The expression and positioning of LC-3 were observed by immunofluorescence staining. TUNEL staining was used to detect cell apoptosis and immunohistochemical analysis was used to evaluate the expression of SP1 in renal tissue after ischemia-reperfusion injury in rats. Results The expression of PTEN was upregulated while SP1 and miR-205 were downregulated in renal ischemia-reperfusion injury. Overexpression of SP1 protected renal tubule cell against injury induced by ischemia-reperfusion via miR-205/PTEN/Akt pathway mediated autophagy. Overexpression of SP1 attenuated renal ischemia-reperfusion injury in rats. Conclusions SP1 overexpression restored autophagy to alleviate acute renal injury induced by ischemia-reperfusion through the miR-205/PTEN/Akt pathway.
Collapse
Affiliation(s)
- Chong Huang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi Province, People's Republic of China
| | - Yan Chen
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi Province, People's Republic of China
| | - Bin Lai
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi Province, People's Republic of China
| | - Yan-Xia Chen
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi Province, People's Republic of China
| | - Cheng-Yun Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi Province, People's Republic of China
| | - Yuan-Fei Liu
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, 330006, Nanchang, Jiangxi Province, People's Republic of China.
| |
Collapse
|
25
|
Terker AS, Sasaki K, Arroyo JP, Niu A, Wang S, Fan X, Zhang Y, Nwosisi S, Zhang MZ, Harris RC. Activation of hypoxia-sensing pathways promotes renal ischemic preconditioning following myocardial infarction. Am J Physiol Renal Physiol 2021; 320:F569-F577. [PMID: 33522414 DOI: 10.1152/ajprenal.00476.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ischemic heart disease is the leading cause of death worldwide and is frequently comorbid with chronic kidney disease. Physiological communication is known to occur between the heart and the kidney. Although primary dysfunction in either organ can induce dysfunction in the other, a clinical entity known as cardiorenal syndrome, mechanistic details are lacking. Here, we used a model of experimental myocardial infarction (MI) to test effects of chronic cardiac ischemia on acute and chronic kidney injury. Surprisingly, chronic cardiac damage protected animals from subsequent acute ischemic renal injury, an effect that was accompanied by evidence of chronic kidney hypoxia. The protection observed post-MI was similar to protection observed in a separate group of healthy animals housed in ambient hypoxic conditions prior to kidney injury, suggesting a common mechanism. There was evidence that chronic cardiac injury activates renal hypoxia-sensing pathways. Increased renal abundance of several glycolytic enzymes following MI suggested that a shift toward glycolysis may confer renal ischemic preconditioning. In contrast, effects on chronic renal injury followed a different pattern, with post-MI animals displaying worsened chronic renal injury and fibrosis. These data show that although chronic cardiac injury following MI protected against acute kidney injury via activation of hypoxia-sensing pathways, it worsened chronic kidney injury. The results further our understanding of cardiorenal signaling mechanisms and have implications for the treatment of heart failure patients with associated renal disease.NEW & NOTEWORTHY Experimental myocardial infarction (MI) protects from subsequent ischemic acute kidney injury but worsens chronic kidney injury. Observed protection from ischemic acute kidney injury after MI was accompanied by chronic kidney hypoxia and increased renal abundance of hypoxia-inducible transcripts. These data support the idea that MI confers protection from renal ischemic injury via chronic renal hypoxia and activation of downstream hypoxia-inducible signaling pathways.
Collapse
Affiliation(s)
- Andrew S Terker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Kensuke Sasaki
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Juan Pablo Arroyo
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Aolei Niu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Suwan Wang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Xiaofeng Fan
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Yahua Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Sochinweichi Nwosisi
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Ming-Zhi Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee
| | - Raymond C Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt Center for Kidney Disease, Nashville, Tennessee.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
26
|
Zuo J, Wang SM, Jiang X, Cao M, Zhang Z, Shi T, Qin HL, Tang W. Design, synthesis and biological evaluation of novel arylpropionic esters for the treatment of acute kidney injury. Bioorg Chem 2020; 105:104455. [PMID: 33197847 DOI: 10.1016/j.bioorg.2020.104455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 12/27/2022]
Abstract
Acute kidney injury (AKI) is associated with a strong inflammatory response, and inhibiting the response effectively prevents or ameliorates AKI. A series of novel arylpropionic esters were designed, synthesized and evaluated their biological activity in LPS-stimulated RAW264.7 cells. Novel arylpropionic esters bearing multi-functional groups showed significant anti-inflammatory activity, in which, compound 13b exhibited the most potent activity through dose-dependent inhibiting the production of nitric oxide (NO, IC50 = 3.52 μM), TNF-α and IL-6 (84.1% and 33.6%, respectively), as well as suppressing the expression of iNOS, COX-2 and TLR4 proteins. In C57BL/6 mice with cisplatin-induced AKI, compound 13b improved kidney function, inhibited inflammatory development, and reduced pathological damage of kidney tissues. In brief, this arylpropionic ester scaffold may be developed as anti-inflammatory agents.
Collapse
Affiliation(s)
- Jiawei Zuo
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China; First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shi-Meng Wang
- School of Life Science, Wuchang University of Technology, Wuhan 430223, China; School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Xia Jiang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China
| | - Mengxin Cao
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China
| | - Ziwen Zhang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China
| | - Tianlu Shi
- First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Hua-Li Qin
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Wenjian Tang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
27
|
Rudman-Melnick V, Adam M, Potter A, Chokshi SM, Ma Q, Drake KA, Schuh MP, Kofron JM, Devarajan P, Potter SS. Single-Cell Profiling of AKI in a Murine Model Reveals Novel Transcriptional Signatures, Profibrotic Phenotype, and Epithelial-to-Stromal Crosstalk. J Am Soc Nephrol 2020; 31:2793-2814. [PMID: 33115917 DOI: 10.1681/asn.2020010052] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Current management of AKI, a potentially fatal disorder that can also initiate or exacerbate CKD, is merely supportive. Therefore, deeper understanding of the molecular pathways perturbed in AKI is needed to identify targets with potential to lead to improved treatment. METHODS We performed single-cell RNA sequencing (scRNA-seq) with the clinically relevant unilateral ischemia-reperfusion murine model of AKI at days 1, 2, 4, 7, 11, and 14 after AKI onset. Using real-time quantitative PCR, immunofluorescence, Western blotting, and both chromogenic and single-molecule in situ hybridizations, we validated AKI signatures in multiple experiments. RESULTS Our findings show the time course of changing gene expression patterns for multiple AKI stages and all renal cell types. We observed elevated expression of crucial injury response factors-including kidney injury molecule-1 (Kim1), lipocalin 2 (Lcn2), and keratin 8 (Krt8)-and of several novel genes (Ahnak, Sh3bgrl3, and Col18a1) not previously examined in kidney pathologies. AKI induced proximal tubule dedifferentiation, with a pronounced nephrogenic signature represented by Sox4 and Cd24a. Moreover, AKI caused the formation of "mixed-identity cells" (expressing markers of different renal cell types) that are normally seen only during early kidney development. The injured tubules acquired a proinflammatory and profibrotic phenotype; moreover, AKI dramatically modified ligand-receptor crosstalk, with potential pathologic epithelial-to-stromal interactions. Advancing age in AKI onset was associated with maladaptive response and kidney fibrosis. CONCLUSIONS The scRNA-seq, comprehensive, cell-specific profiles provide a valuable resource for examining molecular pathways that are perturbed in AKI. The results fully define AKI-associated dedifferentiation programs, potential pathologic ligand-receptor crosstalk, novel genes, and the improved injury response in younger mice, and highlight potential targets of kidney injury.
Collapse
Affiliation(s)
- Valeria Rudman-Melnick
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Andrew Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Saagar M Chokshi
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Meredith P Schuh
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| |
Collapse
|
28
|
Tan RZ, Li JC, Liu J, Lei XY, Zhong X, Wang C, Yan Y, Linda Ye L, Darrel Duan D, Lan HY, Wang L. BAY61-3606 protects kidney from acute ischemia/reperfusion injury through inhibiting spleen tyrosine kinase and suppressing inflammatory macrophage response. FASEB J 2020; 34:15029-15046. [PMID: 32964547 DOI: 10.1096/fj.202000261rrr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) is a highly prevalent clinical syndrome with high mortality and morbidity. Previous studies indicated that inflammation promotes tubular damage and plays a key role in AKI progress. Spleen tyrosine kinase (Syk) has been linked to macrophage-related inflammation in AKI. Up to date, however, no Syk-targeted therapy for AKI has been reported. In this study, we employed both cell model of LPS-induced bone marrow-derived macrophage (BMDM) and mouse model of ischemia/reperfusion injury (IRI)-induced AKI to evaluate the effects of a Syk inhibitor, BAY61-3606 (BAY), on macrophage inflammation in vitro and protection of kidney from AKI in vivo. The expression and secretion of inflammatory cytokines, both in vitro and in vivo, were significantly inhibited even back to normal levels by BAY. The upregulated serum creatinine and blood urea nitrogen levels in the AKI mice were significantly reduced after administration of BAY, implicating a protective effect of BAY on kidneys against IRI. Further analyses from Western blot, immunofluorescence staining and flow cytometry revealed that BAY inhibited the Mincle/Syk/NF-κB signaling circuit and reduced the inflammatory response. BAY also inhibited the reactive oxygen species (ROS), which further decreased the formation of inflammasome and suppressed the mature of IL-1β and IL-18. Notably, these inhibitory effects of BAY on inflammation and inflammasome in BMDM were significantly reversed by Mincle ligand, trehalose-6,6-dibehenate. In summary, these findings provided compelling evidence that BAY may be an efficient inhibitor of the Mincle/Syk/NF-κB signaling circuit and ROS-induced inflammasome, which may help to develop Syk-inhibitors as novel therapeutic agents for AKI.
Collapse
Affiliation(s)
- Rui-Zhi Tan
- Research Center of Traditional Chinese Medicine and Western Medicine Integration, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jian-Chun Li
- Research Center of Traditional Chinese Medicine and Western Medicine Integration, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jian Liu
- Department of Nephrology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xian-Ying Lei
- ICU, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xia Zhong
- Research Center of Traditional Chinese Medicine and Western Medicine Integration, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Chen Wang
- Research Center of Traditional Chinese Medicine and Western Medicine Integration, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ying Yan
- Research Center of Traditional Chinese Medicine and Western Medicine Integration, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Lingyu Linda Ye
- Center for Phenomics of Traditional Chinese Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hui-Yao Lan
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Research Center of Traditional Chinese Medicine and Western Medicine Integration, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
29
|
Funahashi Y, Chowdhury S, Eiwaz MB, Hutchens MP. Acute Cardiorenal Syndrome: Models and Heart-Kidney Connectors. Nephron Clin Pract 2020; 144:629-633. [PMID: 32814315 DOI: 10.1159/000509353] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/10/2020] [Indexed: 11/19/2022] Open
Abstract
Cardiorenal syndrome type 1 (CRS-1) is an acute kidney injury (AKI) due to acute worsening of cardiac function. More than 20% of patients with acute heart failure develop AKI, and AKI predicts poor outcome. Although a number of potential pathways have been suggested as heart-kidney connectors which might drive the syndrome, there are significant barriers to investigation, such as a paucity of animal models, a lack of specific biomarkers, and an inconsistent temporal and causal relationship between changes in cardiac flow and development of renal dysfunction. Thus, mechanisms of heart-kidney interaction are still unclear, and there is no specific or effective therapy for CRS-1. This review, therefore, focuses on mitigating these challenges in the investigation of CRS-1. We review the available models and focus on mechanistic insights gained from those models. In particular, we focus on non-flow and endocrine mediators of CRS-1 such as heart-derived messengers which alter renal function and which may represent targetable pathways in this syndrome. As precise connectors of heart-kidney interaction remain unclear, the establishment of animal and relevant cell-culture models and further investigation are required.
Collapse
Affiliation(s)
- Yoshio Funahashi
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA,
| | - Sheuli Chowdhury
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Mahaba B Eiwaz
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael P Hutchens
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, USA.,Portland Veterans Affairs Medical Center, Operative Care Division, Portland, Oregon, USA
| |
Collapse
|
30
|
Zhang J, Wang X, Wei J, Wang L, Jiang S, Xu L, Qu L, Yang K, Fu L, Buggs J, Cheng F, Liu R. A two-stage bilateral ischemia-reperfusion injury-induced AKI to CKD transition model in mice. Am J Physiol Renal Physiol 2020; 319:F304-F311. [PMID: 32567350 DOI: 10.1152/ajprenal.00017.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) significantly increases the risk of development of chronic kidney disease (CKD). Recently, our laboratory generated a mouse model with the typical phenotypes of AKI to CKD transition in the unilateral kidney. However, AKI, CKD, and even the transition from AKI to CKD usually occur bilaterally rather than unilaterally in patients. Therefore, in the present study, we further modified the strategy and developed a new model of CKD transitioned from bilateral ischemia-reperfusion injury (IRI) in C57BL/6 mice. In this new model, unilateral severe IRI was performed in one kidney while the contralateral kidney was kept intact to maintain animal survival; then, following 14 days of recovery, when the renal function of the injured kidney restored above the survival threshold, the contralateral intact kidney was subjected to a similar IRI. Animals of these two-stage bilateral IRI models with pedicle clamping of 21 and 24 min at a body temperature of 37°C exhibited incomplete recovery from AKI and subsequent development of CKD with characteristics of progressive decline in glomerular filtration rate, increases in plasma creatinine, worsening of proteinuria, and deleterious histopathological changes, including interstitial fibrosis and glomerulosclerosis, in both kidneys. In conclusion, a new bilateral AKI to CKD transition animal model with a typical phenotype of CKD was generated in C57BL/6 mice.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Ximing Wang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Jin Wei
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Shan Jiang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Lan Xu
- College of Public Health, University of South Florida, Tampa, Florida
| | - Larry Qu
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Kun Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Liying Fu
- Tampa General Hospital, Tampa, Florida
| | | | - Feng Cheng
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
31
|
Tourki B, Kain V, Shaikh SR, Leroy X, Serhan CN, Halade GV. Deficit of resolution receptor magnifies inflammatory leukocyte directed cardiorenal and endothelial dysfunction with signs of cardiomyopathy of obesity. FASEB J 2020; 34:10560-10573. [PMID: 32543720 DOI: 10.1096/fj.202000495rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Chronic unresolved inflammation is the primary determinant of cardiovascular disease. Precise mechanisms that define the genesis of unresolved inflammation in heart failure with preserved ejection fraction (HFpEF) are of interest due to the obesity epidemic. To examine the obesity phenotype and its direct/indirect consequences, multiple approaches were employed using the lipoxin receptor (abbreviated as ALX) dysfunction mouse model. Indirect calorimetry analyses revealed that the deletion of ALX dysregulated energy metabolism driving toward age-related obesity. Heart function data suggest that obesity-prone ALX deficient mice had impaired myocardium strain. Comprehensive measurement of chemokines, extracellular matrix, and arrhythmogenic arrays confirmed the dysregulation of multiple ion channels gene expression with amplified inflammatory chemokines and cytokines response at the age of 4 months compared with WT counterparts. Quantitative analyses of leukocytes demonstrated an increase of proinflammatory Ly6Chi CCR2+ macrophages in the spleen and heart at a steady-state resulting in an inflamed splenocardiac axis. Signs of subtle inflammation were marked with cardiorenal, endothelial defects with decreased CD31 and eNOS and an increased iNOS and COX2 expression. Thus, ALX receptor deficiency serves as an experimental model that defines multiple cellular and molecular mechanisms in HFpEF that could be a target for the development of HFpEF therapy in cardiovascular medicine.
Collapse
Affiliation(s)
- Bochra Tourki
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW The current review will describe the current evidence and mechanisms of acute kidney injury (AKI) as a risk factor for long-term kidney complications, summarize the rationale for AKI follow-up and present an approach to monitoring children with AKI. Despite emerging evidence linking AKI with risk for long-term kidney and cardiovascular outcomes, many children who develop AKI are not followed for kidney disease development after hospital discharge. Better understanding of long-term complications after AKI and practical algorithms for follow-up will hopefully increase the rate and quality of post-AKI monitoring. RECENT FINDINGS Recent evidence shows that pediatric AKI is associated with long-term renal outcomes such as chronic kidney disease (CKD) and hypertension, both known to increase cardiovascular risk. The mechanism of AKI progression to CKD involves maladaptive regeneration of tubular epithelial and endothelial cells, inflammation, fibrosis and glomerulosclerosis. Many AKI survivors are not followed, and no guidelines for pediatric AKI follow-up have been published. SUMMARY Children who had AKI are at increased risk of long-term renal complications but many of them are not monitored for these complications. Recognizing long-term outcomes post-AKI and integration of follow-up programs may have a long-lasting positive impact on patient health.
Collapse
|
33
|
Uddin MJ, Dorotea D, Pak ES, Ha H. Fyn Kinase: A Potential Therapeutic Target in Acute Kidney Injury. Biomol Ther (Seoul) 2020; 28:213-221. [PMID: 32336052 PMCID: PMC7216742 DOI: 10.4062/biomolther.2019.214] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/01/2020] [Accepted: 04/06/2020] [Indexed: 01/29/2023] Open
Abstract
Acute kidney injury (AKI) is a common disease with a complex pathophysiology which significantly contributes to the development of chronic kidney disease and end stage kidney failure. Preventing AKI can consequently reduce mortality, morbidity, and healthcare burden. However, there are no effective drugs in use for either prevention or treatment of AKI. Developing therapeutic agents with pleiotropic effects covering multiple pathophysiological pathways are likely to be more effective in attenuating AKI. Fyn, a non-receptor tyrosine kinase, has been acknowledged to integrate multiple injurious stimuli in the kidney. Limited studies have shown increased Fyn transcription level and activation under experimental AKI. Activated Fyn kinase propagates various downstream signaling pathways associated to the progression of AKI, such as oxidative stress, inflammation, endoplasmic reticulum stress, as well as autophagy dysfunction. The versatility of Fyn kinase in mediating various pathophysiological pathways suggests that its inhibition can be a potential strategy in attenuating AKI.
Collapse
Affiliation(s)
- Md Jamal Uddin
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Eun Seon Pak
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
34
|
Terker AS, de Caestecker M. Modeling human disease: a mouse model of acute kidney injury to chronic kidney disease progression after cardiac arrest. Kidney Int 2020; 97:22-24. [PMID: 31901349 PMCID: PMC7197020 DOI: 10.1016/j.kint.2019.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 10/25/2022]
Abstract
Matsushita et al. describe a model of acute kidney injury to chronic kidney disease progression in mice surviving cardiac arrest: mice develop severe acute kidney injury that initially recovers but is followed by the onset of impaired renal function on longer-term follow-up. These findings suggest that distinct cardiorenal toxicities and/or injury dynamics are operative in this cardiac arrest model that do not occur in traditional models of acute kidney injury, providing new opportunities for therapeutic and biomarker discovery for an important clinical problem.
Collapse
Affiliation(s)
- Andrew Scott Terker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mark de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|