101
|
McCullough KR, Akhter J, Taheri MJ, Traylor A, Zmijewska AA, Verma V, Hudson MC, Sachdeva A, Erman EN, Moore KH, George JF, Bolisetty S. Functional consequence of myeloid ferritin heavy chain on acute and chronic effects of rhabdomyolysis-induced kidney injury. Front Med (Lausanne) 2022; 9:894521. [PMID: 36160140 PMCID: PMC9492979 DOI: 10.3389/fmed.2022.894521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Acute kidney injury (AKI) is a serious complication of rhabdomyolysis that significantly impacts survival. Myoglobin released from the damaged muscle accumulates in the kidney, causing heme iron-mediated oxidative stress, tubular cell death, and inflammation. In response to injury, myeloid cells, specifically neutrophils and macrophages, infiltrate the kidneys, and mediate response to injury. Ferritin, comprised of ferritin light chain and ferritin heavy chain (FtH), is vital for intracellular iron handling. Given the dominant role of macrophages and heme-iron burden in the pathogenesis of rhabdomyolysis, we studied the functional role of myeloid FtH in rhabdomyolysis-induced AKI and subsequent fibrosis. Using two models of rhabdomyolysis induced AKI, we found that during the acute phase, myeloid FtH deletion did not impact rhabdomyolysis-induced kidney injury, cell death or cell proliferation, suggesting that tubular heme burden is the dominant injury mechanism. We also determined that, while the kidney architecture was markedly improved after 28 days, tubular casts persisted in the kidneys, suggesting sustained damage or incomplete recovery. We further showed that rhabdomyolysis resulted in an abundance of disparate intra-renal immune cell populations, such that myeloid populations dominated during the acute phase and lymphoid populations dominated in the chronic phase. Fibrotic remodeling was induced in both genotypes at 7 days post-injury but continued to progress only in wild-type mice. This was accompanied by an increase in expression of pro-fibrogenic and immunomodulatory proteins, such as transforming growth factor-β, S100A8, and tumor necrosis factor-α. Taken together, we found that while the initial injury response to heme burden was similar, myeloid FtH deficiency was associated with lesser interstitial fibrosis. Future studies are warranted to determine whether this differential fibrotic remodeling will render these animals more susceptible to a second AKI insult or progress to chronic kidney disease at an accelerated pace.
Collapse
Affiliation(s)
- Kayla R. McCullough
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juheb Akhter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mauhaun J. Taheri
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amie Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anna A. Zmijewska
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vivek Verma
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Matthew C. Hudson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Abhishek Sachdeva
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Elise N. Erman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kyle H. Moore
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James F. George
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Subhashini Bolisetty,
| |
Collapse
|
102
|
Selective EZH2 inhibitor zld1039 alleviates inflammation in cisplatin-induced acute kidney injury partially by enhancing RKIP and suppressing NF-κB p65 pathway. Acta Pharmacol Sin 2022; 43:2067-2080. [PMID: 34937916 PMCID: PMC9343430 DOI: 10.1038/s41401-021-00837-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023]
Abstract
Enhancer of zeste homolog 2 (EZH2), a component of polycomb repressive complex 2 (PRC2), is a histone lysine methyltransferase mediating trimethylation of histone H3 at lysine 27 (H3K27me3), which is a repressive marker at the transcriptional level. EZH2 sustains normal renal function and its overexpression has bad properties. Inhibition of EZH2 overexpression exerts protective effect against acute kidney injury (AKI). A small-molecule compound zld1039 has been developed as an efficient and selective EZH2 inhibitor. In this study, we evaluated the efficacy of zld1039 in the treatment of cisplatin-induced AKI in mice. Before injection of cisplatin (20 mg/kg, i.p.), mice were administered zld1039 (100, 200 mg/kg, i.g.) once, then in the following 3 days. We found that cisplatin-treated mice displayed serious AKI symptoms, evidenced by kidney dysfunction and kidney histological injury, accompanied by EZH2 upregulation in the nucleus of renal tubular epithelial cells. Administration of zld1039 dose-dependently alleviated renal dysfunction as well as the histological injury, inflammation and cell apoptosis in cisplatin-treated mice. We revealed that zld1039 administration exerted an anti-inflammatory effect in kidney of cisplatin-treated mice via H3K27me3 inhibition, raf kinase inhibitor protein (RKIP) upregulation and NF-κB p65 repression. In the cisplatin-treated mouse renal tubular epithelial (TCMK-1) cells, silencing of RKIP with siRNA did not abolish the anti-inflammatory effect of EZH2 inhibition, suggesting that RKIP was partially involved in the anti-inflammatory effect of zld1039. Collectively, EZH2 inhibition alleviates inflammation in cisplatin-induced mouse AKI via upregulating RKIP and blocking NF-κB p65 signaling in cisplatin-induced AKI. The potent and selective EZH2 inhibitor zld1039 has the potential as a promising agent for the treatment of AKI.
Collapse
|
103
|
Jia L, Li C, Bi X, Wei F, Meng J, Sun G, Yu H, Dong H, Li B, Cao Y, Wang L, Jiang A. Prognostic Value of Systemic Immune-Inflammation Index among Critically Ill Patients with Acute Kidney Injury: A Retrospective Cohort Study. J Clin Med 2022; 11:jcm11143978. [PMID: 35887742 PMCID: PMC9319546 DOI: 10.3390/jcm11143978] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 02/05/2023] Open
Abstract
Inflammation plays a significant role in the occurrence and development of acute kidney injury (AKI). Evidence regarding the prognostic effect of the systemic immune-inflammation index (SII) in critically ill patients with AKI is scarce. The aim of this study was to assess the association between SII and all-cause mortality in these patients. Detailed clinical data were extracted from the Medical Information Mart for Intensive Care Database (MIMIC)-IV. The primary outcome was set as the in-hospital mortality. A total of 10,764 AKI patients were enrolled in this study. The restricted cubic splines analyses showed a J-shaped curve between SII and the risk of in-hospital and ICU mortality. After adjusting for relevant confounders, multivariate Cox regression analysis showed that both lower and higher SII levels were associated with an elevated risk of in-hospital all-cause mortality. A similar trend was observed for ICU mortality. In summary, we found that the SII was associated in a J-shaped pattern with all-cause mortality among critically ill patients with AKI. SII appears to be have potential applications in the clinical setting as a novel and easily accessible biomarker for predicting the prognosis of AKI patients.
Collapse
Affiliation(s)
- Lan Jia
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Chen Li
- Department of Orthopaedics, Tianjin Hospital, Tianjin 300211, China;
| | - Xueqing Bi
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Fang Wei
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Jia Meng
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Guijiang Sun
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Haibo Yu
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Hongye Dong
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Bo Li
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Yueqi Cao
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
| | - Lihua Wang
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
- Correspondence: (L.W.); (A.J.); Tel.: +86-022-8832-6796 (L.W.); +86-022-8832-6563 (A.J.)
| | - Aili Jiang
- Department of Kidney Disease and Blood Purification, Institute of Urology & Key Laboratory of Tianjin, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (L.J.); (X.B.); (F.W.); (J.M.); (G.S.); (H.Y.); (H.D.); (B.L.); (Y.C.)
- Correspondence: (L.W.); (A.J.); Tel.: +86-022-8832-6796 (L.W.); +86-022-8832-6563 (A.J.)
| |
Collapse
|
104
|
Li B, Lin F, Xia Y, Ye Z, Yan X, Song B, Yuan T, Li L, Zhou X, Yu W, Cheng F. The Intersection of Acute Kidney Injury and Non-Coding RNAs: Inflammation. Front Physiol 2022; 13:923239. [PMID: 35755446 PMCID: PMC9218900 DOI: 10.3389/fphys.2022.923239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022] Open
Abstract
Acute renal injury (AKI) is a complex clinical syndrome, involving a series of pathophysiological processes, in which inflammation plays a key role. Identification and verification of gene signatures associated with inflammatory onset and progression are imperative for understanding the molecular mechanisms involved in AKI pathogenesis. Non-coding RNAs (ncRNAs), involved in epigenetic modifications of inflammatory responses, are associated with the aberrant expression of inflammation-related genes in AKI. However, its regulatory role in gene expression involves precise transcriptional regulation mechanisms which have not been fully elucidated in the complex and volatile inflammatory response of AKI. In this study, we systematically review current research on the intrinsic molecular mechanisms of ncRNAs that regulate the inflammatory response in AKI. We aim to provide potential research directions and strategies for developing ncRNA-targeted gene therapies as an intervention for the inflammatory damage in AKI.
Collapse
Affiliation(s)
- Bojun Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuqi Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zehua Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinzhou Yan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baofeng Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tianhui Yuan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
105
|
Gao Y, Xu W, Guo C, Huang T. GATA1 regulates the microRNA‑328‑3p/PIM1 axis via circular RNA ITGB1 to promote renal ischemia/reperfusion injury in HK‑2 cells. Int J Mol Med 2022; 50:100. [PMID: 35674159 PMCID: PMC9242654 DOI: 10.3892/ijmm.2022.5156] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022] Open
Abstract
Acute kidney injury (AKI) is caused by renal ischemia/reperfusion injury (IRI) during kidney transplantation. The levels of both circular RNAs (circRNAs) and microRNAs (miRNAs/miR) appear to be critical for AKI detection. While several RNA interactions in AKI have been found, the regulatory mechanisms between the molecules remain to be fully elucidated. In the present study, miRNA expression profiling analysis was conducted using an online dataset to identify the differentially expressed miRNAs in rats with IRI. miR-328-3p was also found to be downregulated in human kidney-2 (HK-2) cells subjected to hypoxia/reperfusion (H/R), and its overexpression targeting pim-1 proto-oncogene (PIM1) resulted in an increased viability and a reduced apoptosis, as well as in the decreased expression of inflammatory factors upon H/R exposure. Putative targets and circRNAs of miR-328-3p were identified using publically available databases. The inhibition of circRNA integrin beta 1 (ITGB1; circITGB1) suppressed the inflammatory response induced by H/R by sponging miR-328-3p in HK-2 cells. Furthermore, a sequence of the functional ITGB1 promoter was studied for transcription factor GATA binding protein 1 (GATA1) binding sites. GATA1 binds to the ITGB1 promoter, leading to the expression of circITGB1. On the whole, the findings of the present study revealed a regulatory pathway modulating miR-328-3p in IRI, demonstrating that the GATA1-mediated regulation of circITGB1 enhanced the H/R-induced inflammatory response via the miR-328-3p/PIM1 axis.
Collapse
Affiliation(s)
- Yang Gao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Weijia Xu
- Department of Kidney Transplantation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Chen Guo
- Department of Kidney Transplantation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Tao Huang
- Department of Kidney Transplantation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
106
|
Zhu Z, Hu J, Chen Z, Feng J, Yang X, Liang W, Ding G. Transition of acute kidney injury to chronic kidney disease: role of metabolic reprogramming. Metabolism 2022; 131:155194. [PMID: 35346693 DOI: 10.1016/j.metabol.2022.155194] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is a global public health concern associated with high morbidity and mortality. Although advances in medical management have improved the in-hospital mortality of severe AKI patients, the renal prognosis for AKI patients in the later period is not encouraging. Recent epidemiological investigations have indicated that AKI significantly increases the risk for the development of chronic kidney disease (CKD) and end-stage renal disease (ESRD) in the future, further contributing to the economic burden on health care systems. The transition of AKI to CKD is complex and often involves multiple mechanisms. Recent studies have suggested that renal tubular epithelial cells (TECs) are more prone to metabolic reprogramming during AKI, in which the metabolic process in the TECs shifts from fatty acid β-oxidation (FAO) to glycolysis due to hypoxia, mitochondrial dysfunction, and disordered nutrient-sensing pathways. This change is a double-edged role. On the one hand, enhanced glycolysis acts as a compensation pathway for ATP production; on the other hand, long-term shut down of FAO and enhanced glycolysis lead to inflammation, lipid accumulation, and fibrosis, contributing to the transition of AKI to CKD. This review discusses developments and therapies focused on the metabolic reprogramming of TECs during AKI, and the emerging questions in this evolving field.
Collapse
Affiliation(s)
- Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Xueyan Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
107
|
Sahu RK, Xavier S, Chauss D, Wang L, Chew C, Taylor R, Stallcup WB, Ma JZ, Kazemian M, Afzali B, Köhl J, Portilla D. Folic acid-mediated fibrosis is driven by C5a receptor 1-mediated activation of kidney myeloid cells. Am J Physiol Renal Physiol 2022; 322:F597-F610. [PMID: 35379003 PMCID: PMC9054266 DOI: 10.1152/ajprenal.00404.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022] Open
Abstract
We have previously reported that increased expression and activation of kidney cell complement components play an important role in the pathogenesis of renal scarring. Here, we used floxed green fluorescent protein (GFP)-C5a receptor 1 (C5aR1) knockin mice (GFP-C5ar1fl/fl) and the model of folic acid (FA)-induced kidney injury to define the cell types and potential mechanisms by which increased C5aR1 activation leads to fibrosis. Using flow cytometry and confocal microscopy, we identified macrophages as the major interstitial cell type showing increased expression of C5aR1 in FA-treated mice. C5ar1fl/fl.Lyz2Cre+/- mice, in which C5aR1 has been specifically deleted in lysozyme M-expressing myeloid cells, experienced reduced fibrosis compared with control C5ar1fl/fl mice. Examination of C5aR1-expressing macrophage transcriptomes by gene set enrichment analysis demonstrated that these cells were enriched in pathways corresponding to the complement cascade, collagen formation, and the NABA matrisome, strongly pointing to their critical roles in tissue repair/scarring. Since C5aR1 was also detected in a small population of platelet-derived growth factor receptor-β+ GFP+ cells, we developed C5ar1fl/fl.Foxd1Cre+/- mice, in which C5aR1 is deleted specifically in pericytes, and found reduced FA-induced fibrosis. Primary cell cultures of platelet-derived growth factor receptor-β+ pericytes isolated from FA-treated C5ar1fl/fl.Foxd1Cre+/- mice showed reduced secretion of several cytokines, including IL-6 and macrophage inflammatory protein-2, compared with pericytes isolated from FA-treated control GFP-C5ar1fl/fl mice. Collectively, these data imply that C5a/C5aR1 axis activation primarily in interstitial cells contributes to the development of renal fibrosis.NEW & NOTEWORTHY This study used novel green fluorescent protein C5a receptor 1 floxed mice and the model of folic acid-mediated kidney fibrosis to demonstrate the pathogenic role of increased expression of this complement receptor on macrophages.
Collapse
Affiliation(s)
- Ranjit K Sahu
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Sandhya Xavier
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, Indiana
| | - Claude Chew
- Flow Cytometry Core, University of Virginia, Charlottesville, Virginia
| | - Ronald Taylor
- Department of Biochemistry, University of Virginia, Charlottesville, Virginia
| | - William B Stallcup
- Sanford Burnham Prebys Medical Discovery Institute, Tumor Microenvironment and Cancer Immunology Program, La Jolla, California
| | - Jennie Z Ma
- Division of Biostatistics, Department of Public Health, University of Virginia, Charlottesville, Virginia
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, Indiana
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Didier Portilla
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
108
|
Xu N, Pang K, Qi S, Wang H. Correlation between perioperative parecoxib use and postoperative acute kidney injury in patients undergoing radical mastectomy: a retrospective cohort analysis. BMC Anesthesiol 2022; 22:155. [PMID: 35596129 PMCID: PMC9121548 DOI: 10.1186/s12871-022-01688-4] [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: 11/25/2021] [Accepted: 05/04/2022] [Indexed: 12/02/2022] Open
Abstract
Background Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most widely prescribed drugs worldwide. However, the effect of NSAIDS on postoperative renal function is still unclear. Few studies have assessed the effects of parecoxib on renal function. Our aim is to investigate a correlation between parecoxib and the presence or absence of AKI postoperatively after a breast cancer surgery operation. Methods This was a retrospective cohort study that we performed on our hospitalized database. From January 2012 to August 2021, 3542 female patients undergoing radical mastectomy were enrolled, all data including the patients' information and laboratory results were obtained from electronic medical system. The main outcome was the incidence of AKI postoperatively. AKI was defined in accordance with the KDIGO criteria. Study groups were treated with or without parecoxib. Univariable and multivariable logistic regression analyses were performed. Results In our study, about 5.76% experienced AKI. The incidence rate of postoperative AKI (3.49%) within 7 days in the parecoxib group was lower than that in the control group (6.00%, P = 0.05). Compared to the control group, the AKI’s incidence was reduced by 49% (OR = 0.46; 95%CI 0.27–0.97) in parecoxib group in multivariable logistic regression analysis. There was a reduction in the incidence of postoperative AKI in other three subgroups: preoperative eGFR < 90 mL/min·1.73/m2 (OR = 0.52; 95%CI 0.27–0.97), blood loss < 1000 ml (OR = 0.48; 95%CI 0.24–0.96) and non-diabetes (OR = 0.51; 95%CI 0.26–0.98). Conclusions Parecoxib was associated with incidence of postoperative acute kidney injury.
Collapse
Affiliation(s)
- Nan Xu
- The Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang, China
| | - Ke Pang
- The Department of Anesthesiology, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Yuelv District, Changsha, Hunan, China
| | - Sihua Qi
- The Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang, China.
| | - Hongmei Wang
- The Department of Pain, The Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, Heilongjiang, China.
| |
Collapse
|
109
|
Yue Y, Yeh JN, Chiang JY, Sung PH, Chen YL, Liu F, Yip HK. Intrarenal arterial administration of human umbilical cord-derived mesenchymal stem cells effectively preserved the residual renal function of diabetic kidney disease in rat. Stem Cell Res Ther 2022; 13:186. [PMID: 35526048 PMCID: PMC9080206 DOI: 10.1186/s13287-022-02857-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/10/2022] [Indexed: 12/16/2022] Open
Abstract
Background This experimental study was designed as a preclinical study for testing the hypothesis that intrarenal arterial (IRA) transfusion of human umbilical cord-derived mesenchymal stem cells (HUCDMSCs) therapy preserved the residual renal function of diabetic kidney disease (DKD) in rat [induction by 5/6 nephrectomy of left kidney and right nephrectomy, followed by intraperitoneal administration of aminoguanidine (180 mg/kg) and streptozotocin (30 mg/kg)]. Methods Animals (n = 24) were categorized into group 1 (sham-operated control), group 2 (DKD), group 3 [DKD + HUCDMSCs (2.1 × 105/IRA injection at day 28 after CKD induction)] and group 4 [(DKD + HUCDMSCs (6.3 × 105/IRA injection)]. Results By day 60 after DKD induction, the kidneys were harvested and the result showed that the creatinine level, ratio of urine protein/urine creatinine and kidney injury score were lowest in group 1, highest in group 2 and significantly lower in group 4 than in group 3 (all p < 0.0001). The protein expressions of apoptotic (cleaved caspase-3/cleaved PARP/mitochondrial Bax), fibrotic (TGF-ß/p-Smad3), autophagic (ratio of LC3B-II/LC3B-I, Atg5/Beclin-1), oxidative stress (NOX-1/NOX-2/oxidized protein/p22phox), mitochondrial/DNA-damaged (cytosolic-cytochrome-C/DRP1/γ-H2AX) and inflammatory (MMP-9/TNF-α/p-NF-κB) biomarkers exhibited an identical pattern, whereas the protein expressions of angiogenesis factors (CD31/vWF/vascularity) exhibited an opposite pattern of creatinine level among the groups (all p < 0.0001). Histopathological findings demonstrated the renal tubular-damaged (KIM-1)/kidney fibrosis area/oxidative stress (8-OHdG + cells) expressed an identical pattern, whereas the podocyte components (ZO-1/synaptopodin/podocin) exhibited an opposite pattern of creatinine level among the groups (all p < 0.0001). No tumorigenesis or immune rejection event was identified. Conclusion IRA injection of xenogeneic MSCs was safe and effectively protected the residual renal function and architectural integrity in DKD rat.
Collapse
Affiliation(s)
- Ya Yue
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Jui-Ning Yeh
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China.,Department of Cardiology, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - John Y Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan.,Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan.,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan.,Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan.,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan.,Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan
| | - Fanna Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China.
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan. .,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan. .,Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan. .,Department of Nursing, Asia University, Taichung, 41354, Taiwan. .,Division of Cardiology, Department of Internal Medicine, Xiamen Chang Gung Hospital, Xiamen, 361028, Fujian, China.
| |
Collapse
|
110
|
|
111
|
Cheung MD, Agarwal A, George JF. Where Are They Now: Spatial and Molecular Diversity of Tissue-Resident Macrophages in the Kidney. Semin Nephrol 2022; 42:151276. [PMID: 36435683 DOI: 10.1016/j.semnephrol.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Kidney resident macrophages (KRMs) are involved in homeostasis, phagocytosis, defense against infectious agents, response to insults, inflammation, and tissue repair. They also play critical roles in the pathogenesis and recovery from many kidney diseases such as acute kidney injury. KRMs historically have been studied as one homogenous population, but the wide-ranging roles and phenotypes observed suggest that there is greater heterogeneity than previously understood. Advancements in RNA sequencing technologies (single-cell RNA sequencing and spatial transcriptomics) have identified specific subsets of KRMs that are molecularly, functionally, and spatially distinct with dynamic changes after kidney injury. Multiple studies have identified unique markers that represent these subpopulations, permitting further characterization of the function and roles they play in the kidney. Understanding the diversity of KRM subpopulations will be key in the development of novel therapies used in treating kidney diseases and promoting kidney health.
Collapse
Affiliation(s)
- Matthew D Cheung
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Department of Veteran Affairs, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - James F George
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama.
| |
Collapse
|
112
|
Sabet N, Soltani Z, Khaksari M. The effects of exercise on kidney injury: the role of SIRT1. Mol Biol Rep 2022; 49:4025-4038. [PMID: 35449317 DOI: 10.1007/s11033-022-07122-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022]
Abstract
In patients with kidney injury, muscle mass and strength decrease with altered muscle protein synthesis and degradation along with complications such as inflammation and low physical activity. A treatment strategy to maintain muscle metabolism in kidney injury is important. One of the proposed strategies in this regard is exercise, which in addition to inducing muscle hypertrophy, reducing plasma creatinine and urea and decreasing the severity of tubal injuries, can boost immune function and has anti-inflammatory effects. One of the molecules that have been considered as a target in the treatment of many diseases is silent information regulator 1 (SIRT1). Exercise increases the expression of SIRT1 and improves its activity. Therefore, studies that examined the effect of exercise on kidney injury considering the role of SIRT1 in this effect were reviewed to determine the direction of kidney injury research in future regarding to its prevalence, especially following diabetes, and lack of definitive treatment. In this review, we found that SIRT1 can be one of renoprotective target pathways of exercise. However, further studies are needed to determine the role of SIRT1 in different kidney injuries following exercise according to the type and severity of exercise, and the type of kidney injury.
Collapse
Affiliation(s)
- Nazanin Sabet
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran.,Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Soltani
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran. .,Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran. .,Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
113
|
Implementation of Hospital-to-Home Model for Nutritional Nursing Management of Patients with Chronic Kidney Disease Using Artificial Intelligence Algorithm Combined with CT Internet. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:1183988. [PMID: 35414801 PMCID: PMC8977294 DOI: 10.1155/2022/1183988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/24/2022]
Abstract
The objective of this study was to evaluate the application value of “Internet + hospital-to-home (H2H)” nutritional care model using the improved wavelet transform algorithm based on computed tomography (CT) images in the nutritional care management of chronic kidney disease (CKD) stages 3-5. A total of 120 patients with CKD were the research objects and they were randomly divided into two groups. The normal nutritional nursing model was used for nursing of patients in the control group, and the “Internet + H2H″ model was used for the observation group (H2H group), with 60 cases in each group. The nursing effect was evaluated using 320-slice volume CT low-dose perfusion imaging images, anthropometry, laboratory biochemical tests, and other survey scores. The results showed that compared with the mean filter denoising (MFD) algorithm and the orthogonal wavelet denoising (OWD) algorithm, the mean square error (MSE) and signal noise ratio (SNR) values of the IWT algorithm were better (40.0781 vs 45.2891, 59.2123)/(20.0122 vs 18.2311, 15.7812) (P < 0.05). The arm muscle circumference (MAC) (239.77 ± 18.24 vs 243.94 ± 18.72 mm) and triceps skindold (TSF) value (8.87 ± 2.74 vs 10.04 ± 2.90 mm) of the patients in the H2H group were greatly improved after the nursing (P < 0.05). For biochemical indicators, serum albumin (ALB) (35.22 ± 4.98 vs 45.32 ± 4.21) g/L, prealbumin (PAB) (289.94 ± 72.99 vs 341.79 ± 74.45) mg/L, hemoglobin (Hb) (97.62 ± 24.87 vs 110.65 ± 28.83) g/L, and blood urea nitrogen (BUN) (15.74 ± 9.87 vs 11.06 ± 5.69) mmol/L of patients in H2H group were improved (P < 0.05). After nursing, the nutritional screening score of the H2H group was obviously improved (83.33% (before) vs 50% (after)), the total score of health quality assessment (114.89 ± 5.23) in the H2H group was much higher than that of the control group (87.22 ± 14.89), and the satisfaction on the nursing model was higher in the H2H group (100% vs 71.67%) (P < 0.05). The renal cortex BF before and after nursing was significantly different between the two groups of patients (P < 0.05), and the BE of the H2H group was significantly higher than that of the control group after treatment ((335.12 ± 52.74) mL·100 g−1·min−1 vs (289.90 ± 53.91) mL·100 g−1·min−1) (P < 0.05). In summary, the “Internet + H2H″ nutritional nursing model was more individualized, which can better improve the physical quality of patients with stages 3-5 of CKD, improve the psychological state of patients, and further enhance the prognosis of the disease. In addition, the IWT algorithm showed better effects in the processing of the image of 320-slice volume CT low-dose perfusion imaging, and it was worthy of clinical application.
Collapse
|
114
|
Hsu CN, Tain YL. Chronic Kidney Disease and Gut Microbiota: What Is Their Connection in Early Life? Int J Mol Sci 2022; 23:3954. [PMID: 35409313 PMCID: PMC9000069 DOI: 10.3390/ijms23073954] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
The gut-kidney interaction implicating chronic kidney disease (CKD) has been the focus of increasing interest in recent years. Gut microbiota-targeted therapies could prevent CKD and its comorbidities. Considering that CKD can originate in early life, its treatment and prevention should start in childhood or even earlier in fetal life. Therefore, a better understanding of how the early-life gut microbiome impacts CKD in later life and how to develop ideal early interventions are unmet needs to reduce CKD. The purpose of the current review is to summarize (1) the current evidence on the gut microbiota dysbiosis implicated in pediatric CKD; (2) current knowledge supporting the impact of the gut-kidney axis in CKD, including inflammation, immune response, alterations of microbiota compositions, short-chain fatty acids, and uremic toxins; and (3) an overview of the studies documenting early gut microbiota-targeted interventions in animal models of CKD of developmental origins. Treatment options include prebiotics, probiotics, postbiotics, etc. To accelerate the transition of gut microbiota-based therapies for early prevention of CKD, an extended comprehension of gut microbiota dysbiosis implicated in renal programming is needed, as well as a greater focus on pediatric CKD for further clinical translation.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
115
|
Olsen E, van Galen G. Chronic Renal Failure-Causes, Clinical Findings, Treatments and Prognosis. Vet Clin North Am Equine Pract 2022; 38:25-46. [DOI: 10.1016/j.cveq.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
116
|
Yao W, Chen Y, Li Z, Ji J, You A, Jin S, Ma Y, Zhao Y, Wang J, Qu L, Wang H, Xiang C, Wang S, Liu G, Bai F, Yang L. Single Cell RNA Sequencing Identifies a Unique Inflammatory Macrophage Subset as a Druggable Target for Alleviating Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103675. [PMID: 35112806 PMCID: PMC9036000 DOI: 10.1002/advs.202103675] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/11/2021] [Indexed: 05/08/2023]
Abstract
Acute kidney injury (AKI) is a complex clinical disorder associated with poor outcomes. Targeted regulation of the degree of inflammation has been a potential strategy for AKI management. Macrophages are the main effector cells of kidney inflammation. However, macrophage heterogeneity in ischemia reperfusion injury induced AKI (IRI-AKI) remains unclear. Using single-cell RNA sequencing of the mononuclear phagocytic system in the murine IRI model, the authors demonstrate the complementary roles of kidney resident macrophages (KRMs) and monocyte-derived infiltrated macrophages (IMs) in modulating tissue inflammation and promoting tissue repair. A unique population of S100a9hi Ly6chi IMs is identified as an early responder to AKI, mediating the initiation and amplification of kidney inflammation. Kidney infiltration of S100A8/A9+ macrophages and the relevance of renal S100A8/A9 to tissue injury is confirmed in human AKI. Targeting the S100a8/a9 signaling with small-molecule inhibitors exhibits renal protective effects represented by improved renal function and reduced mortality in bilateral IRI model, and decreased inflammatory response, ameliorated kidney injury, and improved long-term outcome with decreased renal fibrosis in the unilateral IRI model. The findings support S100A8/A9 blockade as a feasible and clinically relevant therapy potentially waiting for translation in human AKI.
Collapse
Affiliation(s)
- Weijian Yao
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Ying Chen
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Zehua Li
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Jing Ji
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Abin You
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Shanzhao Jin
- Biomedical Pioneering Innovation Center (BIOPIC)Beijing Advanced Innovation Center for Genomics (ICG)School of Life SciencesPeking UniversityBeijing100871China
| | - Yuan Ma
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Youlu Zhao
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Jinwei Wang
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Lei Qu
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Hui Wang
- Laboratory of Electron MicroscopyPathological CenterPeking University First HospitalBeijing100034China
| | - Chengang Xiang
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Suxia Wang
- Laboratory of Electron MicroscopyPathological CenterPeking University First HospitalBeijing100034China
| | - Gang Liu
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC)Beijing Advanced Innovation Center for Genomics (ICG)School of Life SciencesPeking UniversityBeijing100871China
| | - Li Yang
- Renal DivisionPeking University Institute of NephrologyKey Laboratory of Renal Disease‐Ministry of Health of ChinaKey Laboratory of CKD Prevention and Treatment (Peking University)‐Ministry of Education of ChinaResearch Units of Diagnosis and Treatment of Immune‐mediated Kidney Diseases‐Chinese Academy of Medical SciencesPeking University First HospitalXishiku Street #8Beijing100034China
| |
Collapse
|
117
|
Abstract
Acute kidney injury (AKI), characterized by acute renal dysfunction, is an increasingly common clinical problem and an important risk factor in the subsequent development of chronic kidney disease (CKD). Regardless of the initial insults, the progression of CKD after AKI involves multiple types of cells, including renal resident cells and immune cells such as macrophages. Recently, the involvements of macrophages in AKI-to-CKD transition have garnered significant attention. Furthermore, substantial progress has also been made in elucidating the pathophysiological functions of macrophages from the acute kidney to repair or fibrosis. In this review, we highlight current knowledge regarding the roles and mechanisms of macrophage activation and phenotypic polarization, and transdifferentiation in the development of AKI-to-CKD transition. In addition, the potential of macrophage-based therapy for preventing AKI-to-CKD transition is also discussed.
Collapse
|
118
|
Mansour SG, Bhatraju PK, Coca SG, Obeid W, Wilson FP, Stanaway IB, Jia Y, Thiessen-Philbrook H, Go AS, Ikizler TA, Siew ED, Chinchilli VM, Hsu CY, Garg AX, Reeves WB, Liu KD, Kimmel PL, Kaufman JS, Wurfel MM, Himmelfarb J, Parikh SM, Parikh CR. Angiopoietins as Prognostic Markers for Future Kidney Disease and Heart Failure Events after Acute Kidney Injury. J Am Soc Nephrol 2022; 33:613-627. [PMID: 35017169 PMCID: PMC8975075 DOI: 10.1681/asn.2021060757] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/15/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The mechanisms underlying long-term sequelae after AKI remain unclear. Vessel instability, an early response to endothelial injury, may reflect a shared mechanism and early trigger for CKD and heart failure. METHODS To investigate whether plasma angiopoietins, markers of vessel homeostasis, are associated with CKD progression and heart failure admissions after hospitalization in patients with and without AKI, we conducted a prospective cohort study to analyze the balance between angiopoietin-1 (Angpt-1), which maintains vessel stability, and angiopoietin-2 (Angpt-2), which increases vessel destabilization. Three months after discharge, we evaluated the associations between angiopoietins and development of the primary outcomes of CKD progression and heart failure and the secondary outcome of all-cause mortality 3 months after discharge or later. RESULTS Median age for the 1503 participants was 65.8 years; 746 (50%) had AKI. Compared with the lowest quartile, the highest quartile of the Angpt-1:Angpt-2 ratio was associated with 72% lower risk of CKD progression (adjusted hazard ratio [aHR], 0.28; 95% confidence interval [CI], 0.15 to 0.51), 94% lower risk of heart failure (aHR, 0.06; 95% CI, 0.02 to 0.15), and 82% lower risk of mortality (aHR, 0.18; 95% CI, 0.09 to 0.35) for those with AKI. Among those without AKI, the highest quartile of Angpt-1:Angpt-2 ratio was associated with 71% lower risk of heart failure (aHR, 0.29; 95% CI, 0.12 to 0.69) and 68% less mortality (aHR, 0.32; 95% CI, 0.15 to 0.68). There were no associations with CKD progression. CONCLUSIONS A higher Angpt-1:Angpt-2 ratio was strongly associated with less CKD progression, heart failure, and mortality in the setting of AKI.
Collapse
Affiliation(s)
- Sherry G Mansour
- Clinical Translational Research Accelerator, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut.,Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Pavan K Bhatraju
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington.,Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Steven G Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Wassim Obeid
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Francis P Wilson
- Clinical Translational Research Accelerator, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut.,Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Ian B Stanaway
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Yaqi Jia
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | - Alan S Go
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, California.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California.,Division of Nephrology, Department of Medicine, Stanford University, Palo Alto, California.,Department of Health Research and Policy, Stanford University, Palo Alto, California.,Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - T Alp Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Edward D Siew
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Vernon M Chinchilli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Chi-Yuan Hsu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, California.,Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Amit X Garg
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,ICES, Ontario, Canada
| | - W Brian Reeves
- Division of Nephrology, Department of Medicine, University of Texas Joe and Teresa Long School of Medicine, San Antonio, Texas
| | - Kathleen D Liu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, California.,Department of Anesthesia, Division of Critical Care Medicine, University of California, San Francisco, San Francisco, California
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - James S Kaufman
- Division of Nephrology, Veterans Affairs New York Harbor Healthcare System and New York University School of Medicine, New York, New York
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington.,Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Jonathan Himmelfarb
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Samir M Parikh
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | |
Collapse
|
119
|
Wang ZF, Cheng YC, Li YQ, Liu L, Ge SW, Xu G. Characteristics and Prognostic Value of Tertiary Lymphoid Organs in Membranous Nephropathy: A Retrospective Study. Front Med (Lausanne) 2022; 8:803929. [PMID: 35211487 PMCID: PMC8861205 DOI: 10.3389/fmed.2021.803929] [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: 10/28/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Background Tertiary lymphoid organs play an essential role in the inflammation of the kidney. The clinical association between TLOs and membranous nephropathy (MN) is not clear yet. Methods Consecutive patients with the histologically confirmed membranous nephropathy in Tongji Hospital from July 19, 2012, to September 26, 2019, were included in this study. TLOs in renal biopsy tissues were detected by periodic acid–Schiff-stained and immunohistochemistry. Logistic regression was performed to evaluate the correlations of TLOs and clinical features of patients with MN. Kaplan–Meier analysis was utilized to examine the relationship between TLOs and remission of proteinuria. Results A total of 442 patients with MN were included in this study, of which the average age was 46.4 years old, and 58.8% were male. Moreover, 33% of patients with MN had TLOs in this study. The median value of proteinuria among patients with MN with TLOs was 4.9 g/24 h, which was much greater than no-TLOs ones (3.2 g/24 h, p < 0.001). Moreover, the patients with TLOs had higher serum creatinine and lower serum albumin. The severity of clinical features among the patients with MN aggravated with the increase in the grade of TLOs. In addition, the patients who had TLOs were more likely to be positive of anti-phospholipase A2 receptor autoantibodies. Meanwhile, the patients without TLOs showed significantly higher complete remission and total remission of proteinuria. Conclusion In this study, we demonstrated that TLOs were common among patients with MN. Moreover, the patients with MN with TLOs showed a worse clinical manifestation and an outcome compared with the patients without TLOs.
Collapse
Affiliation(s)
- Zu-Feng Wang
- Division of Internal Medicine, Department of Nephrology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Chun Cheng
- Division of Internal Medicine, Department of Nephrology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yue-Qiang Li
- Division of Internal Medicine, Department of Nephrology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Liu
- Division of Internal Medicine, Department of Nephrology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-Wang Ge
- Division of Internal Medicine, Department of Nephrology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Division of Internal Medicine, Department of Nephrology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
120
|
Zheng Z, Tsvetkov D, Bartolomaeus TUP, Erdogan C, Krügel U, Schleifenbaum J, Schaefer M, Nürnberg B, Chai X, Ludwig FA, N'diaye G, Köhler MB, Wu K, Gollasch M, Markó L. Role of TRPC6 in kidney damage after acute ischemic kidney injury. Sci Rep 2022; 12:3038. [PMID: 35194063 PMCID: PMC8864023 DOI: 10.1038/s41598-022-06703-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/03/2022] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential channel subfamily C, member 6 (TRPC6), a non-selective cation channel that controls influx of Ca2+ and other monovalent cations into cells, is widely expressed in the kidney. TRPC6 gene variations have been linked to chronic kidney disease but its role in acute kidney injury (AKI) is unknown. Here we aimed to investigate the putative role of TRPC6 channels in AKI. We used Trpc6-/- mice and pharmacological blockade (SH045 and BI-749327), to evaluate short-term AKI outcomes. Here, we demonstrate that neither Trpc6 deficiency nor pharmacological inhibition of TRPC6 influences the short-term outcomes of AKI. Serum markers, renal expression of epithelial damage markers, tubular injury, and renal inflammatory response assessed by the histological analysis were similar in wild-type mice compared to Trpc6-/- mice as well as in vehicle-treated versus SH045- or BI-749327-treated mice. In addition, we also found no effect of TRPC6 modulation on renal arterial myogenic tone by using blockers to perfuse isolated kidneys. Therefore, we conclude that TRPC6 does not play a role in the acute phase of AKI. Our results may have clinical implications for safety and health of humans with TRPC6 gene variations, with respect to mutated TRPC6 channels in the response of the kidney to acute ischemic stimuli.
Collapse
Affiliation(s)
- Zhihuang Zheng
- Department of Nephrology/Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin, Berlin, Germany.,Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dmitry Tsvetkov
- Department of Nephrology/Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin, Berlin, Germany. .,Department of Geriatrics, University of Greifswald, University District Hospital Wolgast, Greifswald, Germany.
| | - Theda Ulrike Patricia Bartolomaeus
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Cem Erdogan
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ute Krügel
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Johanna Schleifenbaum
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schaefer
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology and Interfaculty Center of Pharmacogenomics and Drug Research, University of Tübingen, Tübingen, Germany
| | - Xiaoning Chai
- Rudolf Boehm Institute for Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Friedrich-Alexander Ludwig
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig, Germany
| | - Gabriele N'diaye
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - May-Britt Köhler
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kaiyin Wu
- Department of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maik Gollasch
- Department of Nephrology/Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin, Berlin, Germany. .,Department of Geriatrics, University of Greifswald, University District Hospital Wolgast, Greifswald, Germany.
| | - Lajos Markó
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin, Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany. .,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
121
|
Molecular Mechanisms of Kidney Injury and Repair. Int J Mol Sci 2022; 23:ijms23031542. [PMID: 35163470 PMCID: PMC8835923 DOI: 10.3390/ijms23031542] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) will become the fifth global cause of death by 2040, thus emphasizing the need to better understand the molecular mechanisms of damage and regeneration in the kidney. CKD predisposes to acute kidney injury (AKI) which, in turn, promotes CKD progression. This implies that CKD or the AKI-to-CKD transition are associated with dysfunctional kidney repair mechanisms. Current therapeutic options slow CKD progression but fail to treat or accelerate recovery from AKI and are unable to promote kidney regeneration. Unraveling the cellular and molecular mechanisms involved in kidney injury and repair, including the failure of this process, may provide novel biomarkers and therapeutic tools. We now review the contribution of different molecular and cellular events to the AKI-to-CKD transition, focusing on the role of macrophages in kidney injury, the different forms of regulated cell death and necroinflammation, cellular senescence and the senescence-associated secretory phenotype (SAPS), polyploidization, and podocyte injury and activation of parietal epithelial cells. Next, we discuss key contributors to repair of kidney injury and opportunities for their therapeutic manipulation, with a focus on resident renal progenitor cells, stem cells and their reparative secretome, certain macrophage subphenotypes within the M2 phenotype and senescent cell clearance.
Collapse
|
122
|
Lu C, Chen H, Wang C, Yang F, Li J, Liu H, Chen G. An Emerging Role of TIM3 Expression on T Cells in Chronic Kidney Inflammation. Front Immunol 2022; 12:798683. [PMID: 35154075 PMCID: PMC8825483 DOI: 10.3389/fimmu.2021.798683] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/16/2021] [Indexed: 12/29/2022] Open
Abstract
T cell immunoglobulin domain and mucin domain 3 (TIM3) was initially identified as an inhibitory molecule on IFNγ-producing T cells. Further research discovered the broad expression of TIM3 on different immune cells binding to multiple ligands. Apart from its suppressive effects on the Th1 cells, recent compelling experiments highlighted the indispensable role of TIM3 in the myeloid cell-mediated inflammatory response, supporting that TIM3 exerts pleiotropic effects on both adaptive and innate immune cells in a context-dependent manner. A large number of studies have been conducted on TIM3 biology in the disease settings of infection, cancer, and autoimmunity. However, there is a lack of clinical evidence to closely evaluate the role of T cell-expressing TIM3 in the pathogenesis of chronic kidney disease (CKD). Here, we reported an intriguing case of Mycobacterium tuberculosis (Mtb) infection that was characterized by persistent overexpression of TIM3 on circulating T cells and ongoing kidney tubulointerstitial inflammation for a period of 12 months. In this case, multiple histopathological biopsies revealed a massive accumulation of recruited T cells and macrophages in the enlarged kidney and liver. After standard anti-Mtb treatment, repeated renal biopsy identified a dramatic remission of the infiltrated immune cells in the tubulointerstitial compartment. This is the first clinical report to reveal a time-course expression of TIM3 on the T cells, which is pathologically associated with the progression of severe kidney inflammation in a non-autoimmunity setting. Based on this case, we summarize the recent findings on TIM3 biology and propose a novel model of CKD progression due to the aberrant crosstalk among immune cells.
Collapse
Affiliation(s)
- Can Lu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huihui Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Chang Wang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Fei Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Jun Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| |
Collapse
|
123
|
Rui-Zhi T, Ke-Huan X, Yuan L, Xiao L, Bing-Wen Z, Tong-Tong L, Li W. Renoprotective effect of isoliquiritigenin on cisplatin-induced acute kidney injury through inhibition of FPR2 in macrophage. J Pharmacol Sci 2022; 148:56-64. [PMID: 34924130 DOI: 10.1016/j.jphs.2021.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is a serious complication in critically ill patients. Accumulating evidences indicated that macrophages play an important pro-inflammatory role in AKI and isoliquiritigenin (ISL) can inhibit macrophagic inflammation, but its role in AKI and the underlying mechanism are unknown. The present study aims to investigate the renoprotective effect of ISL on AKI and the role of Formyl peptide receptors 2 (FPR2) in this process. In this study, cisplatin-induced AKI model and lipopolysaccharide-induced macrophage inflammatory model were employed to perform the in vivo and in vitro experiments. The results showed that ISL strongly relieved kidney injury and inhibited renal inflammation in vivo and suppress macrophagic inflammatory response in vitro. Importantly, it was found that FPR2 was significantly upregulated compared to the control group in AKI and LPS-induced macrophage, whereas it was strongly suppressed by ISL. Interestingly, overexpression of FPR2 with transfection of pcDNA3.1-FPR2 effectively reversed the anti-inflammatory effect of ISL in macrophage, suggesting that FPR2 may be the potential target for ISL to prevent inflammation and improve kidney injury of AKI. Take together, these findings indicated that ISL improved cisplantin-induced kidney injury by inhibiting FPR2 involved macrophagic inflammation, which may provide a potential therapeutic option for AKI.
Collapse
MESH Headings
- Acute Kidney Injury/chemically induced
- Acute Kidney Injury/drug therapy
- Acute Kidney Injury/genetics
- Acute Kidney Injury/prevention & control
- Animals
- Cells, Cultured
- Chalcones/isolation & purification
- Chalcones/pharmacology
- Chalcones/therapeutic use
- Cisplatin/adverse effects
- Gene Expression/drug effects
- Glycyrrhiza/chemistry
- Inflammation
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Molecular Targeted Therapy
- Phytotherapy
- Receptors, Formyl Peptide/antagonists & inhibitors
- Receptors, Formyl Peptide/genetics
- Receptors, Formyl Peptide/metabolism
- Receptors, Formyl Peptide/physiology
- Receptors, Lipoxin/antagonists & inhibitors
- Receptors, Lipoxin/genetics
- Receptors, Lipoxin/metabolism
- Receptors, Lipoxin/physiology
- Up-Regulation/drug effects
- Mice
Collapse
Affiliation(s)
- Tan Rui-Zhi
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xie Ke-Huan
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Liao Yuan
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lin Xiao
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhu Bing-Wen
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Liu Tong-Tong
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wang Li
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
124
|
Chen T, Jiang Y, Xu S, Cheuk YC, Wang J, Yang C, Rong R. Poly(I:C)-Induced Mesenchymal Stem Cells Protect the Kidney Against Ischemia/Reperfusion Injury via the TLR3/PI3K Pathway. Front Med (Lausanne) 2021; 8:755849. [PMID: 34901066 PMCID: PMC8655722 DOI: 10.3389/fmed.2021.755849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/04/2021] [Indexed: 11/24/2022] Open
Abstract
Objective: To investigate the effect and protective mechanism of mesenchymal stem cell subpopulations on acute kidney injury by establishing a mouse model of renal ischemia-reperfusion injury. Methods: Male C57BL/6 mice were randomly divided into five groups, namely, sham-operation group and those treated with normal saline, untreated mesenchymal stem cells, mesenchymal stem cells treated with lipopolysaccharide (LPS, pro-inflammatory phenotype) and mesenchymal stem cells treated with polyinosinic-polycytidylic acid (poly[I:C], anti-inflammatory phenotype) respectively. The renal function, histopathological damage, circulating inflammation levels and antioxidant capacity of mice were evaluated. The PI3 kinase p85 (PI3K) inhibitor was added into the conventional mesenchymal stem cell cultures in vitro to observe its effects on the secretion of anti-inflammatory cytokines. Results: Mesenchymal stem cells treated with poly(I:C) (anti-inflammatory phenotype) could effectively reduce serum creatinine and blood urea nitrogen, attenuate histopathological damage and apoptosis level, decrease the level of circulating pro-inflammatory cytokines and increase the level of circulating anti-inflammatory cytokines, enhance peroxidase activity and reduce malondialdehyde content at each time point. After the addition of the PI3K inhibitor, the mRNA expression and protein secretion of indoleamine 2,3-dioxygenase 1 and heme oxygenase 1 of various mesenchymal stem cells were significantly reduced, and that of mesenchymal stem cells treated with poly(I:C) (anti-inflammatory phenotype) was more obvious. Conclusions: Polyriboinosinic-polyribocytidylic acid (poly[I:C]), a synthetic double-stranded RNA, whose pretreatment induces mesenchymal stem cells to differentiate into the anti-inflammatory phenotype. Anti-inflammatory mesenchymal stem cells induced by poly(I:C) can better protect renal function, alleviate tissue damage, reduce circulating inflammation levels and enhance antioxidant capacity, and achieve stronger anti-inflammatory effects through the TLR3/PI3K pathway.
Collapse
Affiliation(s)
- Tian Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yamei Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Shihao Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jiyan Wang
- Department of Urology, Shanghai Public Health Clinical Center, Shanghai, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
125
|
Assessing and counteracting fibrosis is a cornerstone of the treatment of CKD secondary to systemic and renal limited autoimmune disorders. Autoimmun Rev 2021; 21:103014. [PMID: 34896651 DOI: 10.1016/j.autrev.2021.103014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is an increasing cause of morbidity and mortality worldwide. Besides the higher prevalence of diabetes, hypertension and aging worldwide, immune mediated disorders remain an important cause of kidney disease and are especially prevalent in young adults. Regardless of the initial insult, final pathway to CKD and kidney failure is always the loss of normal tissue and fibrosis development, in which the dynamic equilibrium between extracellular matrix synthesis and degradation is disturbed, leading to excessive production and accumulation. During fibrosis, a multitude of cell types intervene at different levels, but myofibroblasts and inflammatory cells are considered critical in the process. They exert their effects through different molecular pathways, of which transforming growth factor β (TGF-β) has demonstrated to be of particular importance. Additionally, CKD itself promotes fibrosis due to the accumulation of toxins and hormonal changes, and proteinuria is simultaneously a manifestation of CKD and a specific driver of renal fibrosis. Pathways involved in renal fibrosis and CKD are closely interrelated, and although important advances have been made in our knowledge of them, it is still necessary to translate them into clinical practice. Given the complexity of this process, it is highly likely that its treatment will require a multi-target strategy to control the origin of the damage but also the mechanisms that perpetuate it. Fortunately, rapid technology development over the last years and new available drugs in the nephrologist's armamentarium give reasons for optimism that more personalized assistance for CKD and renal fibrosis will appear in the future.
Collapse
|
126
|
Sato Y, Oguchi A, Fukushima Y, Masuda K, Toriu N, Taniguchi K, Yoshikawa T, Cui X, Kondo M, Hosoi T, Komidori S, Shimizu Y, Fujita H, Jiang L, Kong Y, Yamanashi T, Seita J, Yamamoto T, Toyokuni S, Hamazaki Y, Hattori M, Yoshikai Y, Boor P, Floege J, Kawamoto H, Murakawa Y, Minato N, Yanagita M. CD153-CD30 signaling promotes age-dependent tertiary lymphoid tissue expansion and kidney injury. J Clin Invest 2021; 132:146071. [PMID: 34813503 PMCID: PMC8759786 DOI: 10.1172/jci146071] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Tertiary lymphoid tissues (TLTs) facilitate local T and B cell interactions in chronically inflamed organs. However, the cells and molecular pathways that govern TLT formation are poorly defined. Here, we identified TNF superfamily CD153/CD30 signaling between 2 unique age-dependent lymphocyte subpopulations, CD153+PD-1+CD4+ senescence-associated T (SAT) cells and CD30+T-bet+ age-associated B cells (ABCs), as a driver for TLT expansion. SAT cells, which produced ABC-inducing factors IL-21 and IFN-γ, and ABCs progressively accumulated within TLTs in aged kidneys after injury. Notably, in kidney injury models, CD153 or CD30 deficiency impaired functional SAT cell induction, which resulted in reduced ABC numbers and attenuated TLT formation with improved inflammation, fibrosis, and renal function. Attenuated TLT formation after transplantation of CD153-deficient bone marrow further supported the importance of CD153 in immune cells. Clonal analysis revealed that SAT cells and ABCs in the kidneys arose from both local differentiation and recruitment from the spleen. In the synovium of aged rheumatoid arthritis patients, T peripheral helper/T follicular helper cells and ABCs also expressed CD153 and CD30, respectively. Together, our data reveal a previously unappreciated function of CD153/CD30 signaling in TLT formation and propose targeting the CD153/CD30 signaling pathway as a therapeutic target for slowing kidney disease progression.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akiko Oguchi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Fukushima
- Department of Immunosenescence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kyoko Masuda
- Department of Immunology, Institute for Frontier Medical Science, Kyoto University, Kyoto, Japan
| | - Naoya Toriu
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keisuke Taniguchi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahisa Yoshikawa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Xiaotong Cui
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makiko Kondo
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Hosoi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shota Komidori
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoko Shimizu
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Harumi Fujita
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Li Jiang
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yingyi Kong
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Jun Seita
- Medical Sciences Innovation Hub Program, RIKEN, Tokyo, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Hamazaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masakazu Hattori
- Department of Immunosenescence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasunobu Yoshikai
- Division of Host Defense, Network Center for Infectious Disease, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Peter Boor
- Department of Nephrology, University Hospital RWTH Aachen, Aachen, Germany
| | - Jürgen Floege
- Department of Nephrology, University Hospital RWTH Aachen, Aachen, Germany
| | - Hiroshi Kawamoto
- Department of Immunology, Institute for Frontier Medical Science, Kyoto University, Kyoto, Japan
| | - Yasuhiro Murakawa
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Nagahiro Minato
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
127
|
Baranwal G, Creed HA, Black LM, Auger A, Quach AM, Vegiraju R, Eckenrode HE, Agarwal A, Rutkowski JM. Expanded renal lymphatics improve recovery following kidney injury. Physiol Rep 2021; 9:e15094. [PMID: 34806312 PMCID: PMC8606868 DOI: 10.14814/phy2.15094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Acute kidney injury (AKI) is a major cause of patient mortality and a major risk multiplier for the progression to chronic kidney disease (CKD). The mechanism of the AKI to CKD transition is complex but is likely mediated by the extent and length of the inflammatory response following the initial injury. Lymphatic vessels help to maintain tissue homeostasis through fluid, macromolecule, and immune modulation. Increased lymphatic growth, or lymphangiogenesis, often occurs during inflammation and plays a role in acute and chronic disease processes. What roles renal lymphatics and lymphangiogenesis play in AKI recovery and CKD progression remains largely unknown. To determine if the increased lymphatic density is protective in the response to kidney injury, we utilized a transgenic mouse model with inducible, kidney-specific overexpression of the lymphangiogenic protein vascular endothelial growth factor-D to expand renal lymphatics. "KidVD" mouse kidneys were injured using inducible podocyte apoptosis and proteinuria (POD-ATTAC) or bilateral ischemia reperfusion. In the acute injury phase of both models, KidVD mice demonstrated a similar loss of function measured by serum creatinine and glomerular filtration rate compared to their littermates. While the initial inflammatory response was similar, KidVD mice demonstrated a shift toward more CD4+ and fewer CD8+ T cells in the kidney. Reduced collagen deposition and improved functional recovery over time was also identified in KidVD mice. In KidVD-POD-ATTAC mice, an increased number of podocytes were counted at 28 days post-injury. These data demonstrate that increased lymphatic density prior to injury alters the injury recovery response and affords protection from CKD progression.
Collapse
Affiliation(s)
- Gaurav Baranwal
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Heidi A. Creed
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Laurence M. Black
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Nephrology Research and Training CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Alexa Auger
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Alexander M. Quach
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Rahul Vegiraju
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Han E. Eckenrode
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Nephrology Research and Training CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Anupam Agarwal
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Nephrology Research and Training CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Department of Veterans AffairsBirmingham Veterans Administration Medical CenterBirminghamAlabamaUSA
| | - Joseph M. Rutkowski
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| |
Collapse
|
128
|
NLRP3 associated with chronic kidney disease progression after ischemia/reperfusion-induced acute kidney injury. Cell Death Discov 2021; 7:324. [PMID: 34716316 PMCID: PMC8556399 DOI: 10.1038/s41420-021-00719-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 11/08/2022] Open
Abstract
Nod-like receptor protein 3 (NLRP3), as an inflammatory regulator, has been implicated in acute kidney injury (AKI). Failed recovery after AKI can lead to chronic kidney disease (CKD). However, the role of NLRP3 in the AKI-CKD transition is still unknown. A mild or severe AKI mouse model was performed by using ischemia-reperfusion injury (IRI). We evaluated the renal NLRP3 expression in acute and chronic phases of ischemic AKI, respectively. Although serum creatinine (Cr) and blood urea nitrogen (BUN) levels in AKI chronic phase were equivalent to normal baseline, histological analysis and fibrotic markers revealed that severe AKI-induced maladaptive tubular repair with immune cell infiltration and fibrosis. Tubular damage was restored completely in mild AKI rather than in severe AKI. Of note, persistent overexpression of NLRP3 was also found in severe AKI but not in mild AKI. In the severe AKI-induced chronic phase, there was a long-term high level of NLRP3 in serum or urine. Overt NLRP3 was mainly distributed in the abnormal tubules surrounded by inflammatory infiltrates and fibrosis, which indicated the maladaptive repair. Renal Nlrp3 overexpression was correlated with infiltrating macrophages and fibrosis. Renal NLRP3 signaling-associated genes were upregulated after severe AKI by RNA-sequencing. Furthermore, NLRP3 was found increased in renal tubular epitheliums from CKD biopsies. Together, persistent NLRP3 overexpression was associated with chronic pathological changes following AKI, which might be a new biomarker for evaluating the possibility of AKI-CKD transition.
Collapse
|
129
|
Wu YM, Shi Q, Zhu PF, Ma HJ, Cui SC, Li J, Hou AJ, Li JY. Rhodomeroterpene alleviates macrophage infiltration and the inflammatory response in renal tissue to improve acute kidney injury. FASEB J 2021; 35:e21985. [PMID: 34674317 DOI: 10.1096/fj.202100981rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022]
Abstract
Inflammation is broadly recognized as an important factor in the pathogenesis of acute kidney injury (AKI), but pharmacological approaches to alleviate inflammation in AKI have not been proved successful in clinical trials. Macrophage infiltration into renal tissue promotes inflammatory responses that contribute to the pathogenesis of AKI. Suppression of renal tissue inflammatory responses is postulated to improve renal injury of patients and animals. Rhodomeroterpene (RMT) is a novel meroterpenoid isolated from the Rhododendron genus that was shown to exert anti-inflammatory action in vivo or in vitro in this study. We investigated the treatment effects of RMT on LPS-induced sepsis and two different AKI models. The results showed that pretreatment with RMT (30 mg kg-1 d-1 , ip, for 3 days) significantly inhibited acute inflammatory responses in LPS-induced septic mice. In both renal ischemia-reperfusion injury (I/R) and sepsis-induced AKI models, RMT (30 mg kg-1 d-1 , ip, for 3 days) ameliorated renal function and injury and alleviated inflammation by reducing the infiltration of immune cells, including macrophages and neutrophils. Furthermore, our study demonstrated that RMT inhibits inflammatory responses in macrophages. The anti-inflammatory effects of RMT may be due to the inactivation of the IKK/NF-κB and PI3K/PDK1/Akt inflammatory signaling pathways in macrophages. Collectively, our findings indicate that RMT ameliorates renal injury and alleviates the renal inflammatory state in different AKI models, suggesting that RMT may be a potential agent for the treatment of AKI.
Collapse
Affiliation(s)
- Yong-Mei Wu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qing Shi
- School of Pharmacy, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Peng-Fei Zhu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hai-Jian Ma
- School of Pharmacy, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Shi-Chao Cui
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jia Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ai-Jun Hou
- School of Pharmacy, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Jing-Ya Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
130
|
Shao L, Ma Y, Fang Q, Huang Z, Wan S, Wang J, Yang L. Role of protein phosphatase 2A in kidney disease (Review). Exp Ther Med 2021; 22:1236. [PMID: 34539832 PMCID: PMC8438693 DOI: 10.3892/etm.2021.10671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Kidney disease affects millions of people worldwide and is a financial burden on the healthcare system. Protein phosphatase 2A (PP2A), which is involved in renal development and the function of ion-transport proteins, aquaporin-2 and podocytes, is likely to serve an important role in renal processes. PP2A is associated with the pathogenesis of a variety of different kidney diseases including podocyte injury, inflammation, tumors and chronic kidney disease. The current review aimed to discuss the structure and function of PP2A subunits in the context of kidney diseases. How dysregulation of PP2A in the kidneys causes podocyte death and the inactivation of PP2A in renal carcinoma tissues is discussed. Inhibition of PP2A activity prevents epithelial-mesenchymal transition and attenuates renal fibrosis, creating a favorable inflammatory microenvironment and promoting the initiation and progression of tumor pathogenesis. The current review also indicates that PP2A serves an important role in protection against renal inflammation. Understanding the detailed mechanisms of PP2A provides information that can be utilized in the design and application of novel therapeutics for the treatment and prevention of renal diseases.
Collapse
Affiliation(s)
- Lishi Shao
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Yiqun Ma
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Qixiang Fang
- Department of Urology, The First Affiliated Hospital of the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P.R. China
| | - Ziye Huang
- Department of Urology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Shanshan Wan
- Department of Radiology, Yunnan Kun-Gang Hospital, Anning, Yunnan 650300, P.R. China
| | - Jiaping Wang
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Li Yang
- Department of Anatomy, Histology and Embryology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
131
|
Yan MT, Chao CT, Lin SH. Chronic Kidney Disease: Strategies to Retard Progression. Int J Mol Sci 2021; 22:ijms221810084. [PMID: 34576247 PMCID: PMC8470895 DOI: 10.3390/ijms221810084] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD), defined as the presence of irreversible structural or functional kidney damages, increases the risk of poor outcomes due to its association with multiple complications, including altered mineral metabolism, anemia, metabolic acidosis, and increased cardiovascular events. The mainstay of treatments for CKD lies in the prevention of the development and progression of CKD as well as its complications. Due to the heterogeneous origins and the uncertainty in the pathogenesis of CKD, efficacious therapies for CKD remain challenging. In this review, we focus on the following four themes: first, a summary of the known factors that contribute to CKD development and progression, with an emphasis on avoiding acute kidney injury (AKI); second, an etiology-based treatment strategy for retarding CKD, including the approaches for the common and under-recognized ones; and third, the recommended approaches for ameliorating CKD complications, and the final section discusses the novel agents for counteracting CKD progression.
Collapse
Affiliation(s)
- Ming-Tso Yan
- Department of Medicine, Division of Nephrology, Cathay General Hospital, School of Medicine, Fu-Jen Catholic University, Taipei 106, Taiwan;
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei 114, Taiwan
| | - Chia-Ter Chao
- Department of Internal Medicine, Nephrology Division, National Taiwan University Hospital, Taipei 104, Taiwan;
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 104, Taiwan
- Department of Internal Medicine, Nephrology Division, National Taiwan University College of Medicine, Taipei 104, Taiwan
| | - Shih-Hua Lin
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei 114, Taiwan
- Department of Internal Medicine, Nephrology Division, National Defense Medical Center, Taipei 104, Taiwan
- Correspondence: or
| |
Collapse
|
132
|
Kraińska MM, Pietrzkowska N, Turlej E, Zongjin L, Marycz K. Extracellular vesicles derived from mesenchymal stem cells as a potential therapeutic agent in acute kidney injury (AKI) in felines: review and perspectives. Stem Cell Res Ther 2021; 12:504. [PMID: 34526105 PMCID: PMC8444608 DOI: 10.1186/s13287-021-02573-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/27/2021] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs), known from their key role in the regeneration process of tissues, and their abilities to release bioactive factors like extracellular vesicles (EVs) could be considered as a potential, modern tool in the treatment of AKI (acute kidney injury) in both human and veterinary patients. The complex pathophysiology of a renal function disorder (AKI) makes difficult to find a universal therapy, but the treatment strategy is based on MSCs and derived from them, EVs seem to solve this problem. Due to their small size, the ability of the cargo transport, the ease of crossing the barriers and the lack of the ability to proliferate and differentiate, EVs seem to have a significant impact on the development such therapy. Their additional impact associated with their ability to modulate immune response and inflammation process, their strong anti-fibrotic and anti-apoptotic effects and the relation with the releasing of the reactive oxygen species (ROS), that pivotal role in the AKI development is undoubtedly, limits the progress of AKI. Moreover, the availability of EVs from different sources encourages to extend research with using EVs from MSCs in AKI treatment in felines; in that, the possibilities of kidney injuries treatment are still limited to the classical therapies burdened with dangerous side effects. In this review, we underline the significance of the processes, in whose EVs are included during the AKI in order to show the potential benefits of EVs-MSCs-based therapies against AKI in felines.
Collapse
Affiliation(s)
- Magdalena M Kraińska
- International Institute of Translational Medicine (MIMT), Jesionowa St 11, 55-114, Malin, Wisznia Mała, Poland.,Department of General and Transplant Surgery, Warsaw Medical University, Nowogrodzka St 59, 02-014, Warszawa, Poland
| | - Natalia Pietrzkowska
- International Institute of Translational Medicine (MIMT), Jesionowa St 11, 55-114, Malin, Wisznia Mała, Poland
| | - Eliza Turlej
- Department of Experimental Biology, Wroclaw University of Environmental and Life Science, Norwida St 27B, 50-375, Wrocław, Poland
| | - Li Zongjin
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China
| | - Krzysztof Marycz
- International Institute of Translational Medicine (MIMT), Jesionowa St 11, 55-114, Malin, Wisznia Mała, Poland. .,Department of Experimental Biology, Wroclaw University of Environmental and Life Science, Norwida St 27B, 50-375, Wrocław, Poland.
| |
Collapse
|
133
|
Wang CY, Chen WL, Cheng-Chung Wei J. Link Between Inflammatory Bowel Disease and Risk of Chronic Kidney Disease. Clin Gastroenterol Hepatol 2021; 19:1993-1994. [PMID: 33248103 DOI: 10.1016/j.cgh.2020.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Cheng-You Wang
- Department of Medicine, E-DA Hospital, Kaohsiung, Taiwan
| | - Wei-Liang Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - James Cheng-Chung Wei
- Department of Rheumatology & Immunology, Chung Shan Medical University Hospital; Institute of Medicine, Chung Shan Medical University; Graduate Institute of Integrated Medicine. China Medical University, Taichung, Taiwan
| |
Collapse
|
134
|
Interplay between extracellular matrix components and cellular and molecular mechanisms in kidney fibrosis. Clin Sci (Lond) 2021; 135:1999-2029. [PMID: 34427291 DOI: 10.1042/cs20201016] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is characterized by pathological accumulation of extracellular matrix (ECM) proteins in renal structures. Tubulointerstitial fibrosis is observed in glomerular diseases as well as in the regeneration failure of acute kidney injury (AKI). Therefore, finding antifibrotic therapies comprises an intensive research field in Nephrology. Nowadays, ECM is not only considered as a cellular scaffold, but also exerts important cellular functions. In this review, we describe the cellular and molecular mechanisms involved in kidney fibrosis, paying particular attention to ECM components, profibrotic factors and cell-matrix interactions. In response to kidney damage, activation of glomerular and/or tubular cells may induce aberrant phenotypes characterized by overproduction of proinflammatory and profibrotic factors, and thus contribute to CKD progression. Among ECM components, matricellular proteins can regulate cell-ECM interactions, as well as cellular phenotype changes. Regarding kidney fibrosis, one of the most studied matricellular proteins is cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), currently considered as a fibrotic marker and a potential therapeutic target. Integrins connect the ECM proteins to the actin cytoskeleton and several downstream signaling pathways that enable cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. In kidney fibrosis, there is an increase in ECM deposition, lower ECM degradation and ECM proteins cross-linking, leading to an alteration in the tissue mechanical properties and their responses to injurious stimuli. A better understanding of these complex cellular and molecular events could help us to improve the antifibrotic therapies for CKD.
Collapse
|
135
|
Ishiuchi N, Nakashima A, Doi S, Kanai R, Maeda S, Takahashi S, Nagao M, Masaki T. Serum-free medium and hypoxic preconditioning synergistically enhance the therapeutic effects of mesenchymal stem cells on experimental renal fibrosis. Stem Cell Res Ther 2021; 12:472. [PMID: 34425892 PMCID: PMC8381539 DOI: 10.1186/s13287-021-02548-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/08/2021] [Indexed: 01/27/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) repair injured tissue in a paracrine manner. To enhance their therapeutic properties, preconditioning with various factors has been researched. We have previously showed that MSCs cultured in serum-free medium (SF-MSCs) promote their immunosuppressive ability, thereby enhancing their anti-fibrotic effect. Here, we examined whether serum-free medium and hypoxic preconditioning synergistically enhance the therapeutic effects of MSCs on renal fibrosis in rats with ischemia–reperfusion injury (IRI). Methods SF-MSCs were incubated under 1% O2 conditions (hypo-SF-MSCs) or 21% O2 conditions (normo-SF-MSCs) for 24 h before collection. After IRI procedure, hypo-SF-MSCs or normo-SF-MSCs were injected through the abdominal aorta. At 7 or 21 days post-injection, the rats were killed and their kidneys were collected to evaluate inflammation and fibrosis. In in vitro experiments, we investigated whether hypo-SF-MSCs enhanced secretion of anti-fibrotic humoral factors using transforming growth factor (TGF)-β1-stimulated HK-2 cells incubated with conditioned medium from hypo-SF-MSCs or normo-SF-MSCs. Results Normo-SF-MSCs showed attenuation of senescence, which increased their proliferative capacity. Although no significant difference in cellular senescence was found between normo-SF-MSCs and hypo-SF-MSCs, hypo-SF-MSCs further increased their proliferative capacity compared with normo-SF-MSCs. Additionally, administration of hypo-SF-MSCs more strongly ameliorated renal fibrosis than that of normo-SF-MSCs. Moreover, although hypo-SF-MSCs strongly attenuated infiltration of inflammatory cells compared with the control rats, which were treated with PBS, this attenuation was almost equal between normo-SF-MSCs and hypo-SF-MSCs. In vitro experiments revealed that hypo-SF-MSCs more significantly inhibited transforming growth factor (TGF)-β/Smad signaling compared with normo-SF-MSCs. Moreover, hypoxic preconditioning increased hepatocyte growth factor (HGF) secretion even under serum-free conditions, whereas knockdown of HGF in hypo-SF-MSCs attenuated inhibition of TGF-β/Smad signaling. Conclusions These results indicate that administration of ex vivo-expanded, hypoxia-preconditioned SF-MSCs may be a useful cell therapy to prevent renal fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02548-7.
Collapse
Affiliation(s)
- Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan.,Center for Cause of Death Investigation Research, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.,Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan. .,Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Shigehiro Doi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Ryo Kanai
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Satoshi Maeda
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.,TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Shinya Takahashi
- Department of Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Masataka Nagao
- Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan.
| |
Collapse
|
136
|
Zheng Z, Li C, Shao G, Li J, Xu K, Zhao Z, Zhang Z, Liu J, Wu H. Hippo-YAP/MCP-1 mediated tubular maladaptive repair promote inflammation in renal failed recovery after ischemic AKI. Cell Death Dis 2021; 12:754. [PMID: 34330891 PMCID: PMC8324794 DOI: 10.1038/s41419-021-04041-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Acute kidney injury (AKI) is associated with significant morbidity and its chronic inflammation contributes to subsequent chronic kidney disease (CKD) development. Yes-associated protein (YAP), the major transcriptional coactivator of the Hippo pathway, has been shown associated with chronic inflammation, but its role and mechanism in AKI-CKD transition remain unclear. Here we aimed to investigate the role of YAP in AKI-induced chronic inflammation. Renal ischemia/reperfusion (I/R) was used to induce a mouse model of AKI-CKD transition. We used verteporfin (VP), a pharmacological inhibitor of YAP, to treat post-IRI mice for a period, and evaluated the influence of YAP inhibition on long-term outcomes of AKI. In our results, severe IRI led to maladaptive tubular repair, macrophages infiltration, and progressive fibrosis. Following AKI, the Hippo pathway was found significantly altered with YAP persistent activation. Besides, tubular YAP activation was associated with the maladaptive repair, also correlated with interstitial macrophage infiltration. Monocyte chemoattractant protein 1 (MCP-1) was found notably upregulated with YAP activation. Of note, pharmacological inhibition of YAP in vivo attenuated renal inflammation, including macrophage infiltration and MCP-1 overexpression. Consistently, in vitro oxygen-glucose deprivation and reoxygenation (OGD/R) induced YAP activation and MCP-1 overproduction whereas these could be inhibited by VP. In addition, we modulated YAP activity by RNA interference, which further confirmed YAP activation enhances MCP-1 expression. Together, we concluded tubular YAP activation with maladaptive repair exacerbates renal inflammation probably via promoting MCP-1 production, which contributes to AKI-CKD transition.
Collapse
Affiliation(s)
- Zhihuang Zheng
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chuanlei Li
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guangze Shao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jinqing Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kexin Xu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhigang Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jun Liu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
137
|
Lu HY, Wang GY, Zhao JW, Jiang HT. Knockdown of lncRNA MALAT1 ameliorates acute kidney injury by mediating the miR-204/APOL1 pathway. J Clin Lab Anal 2021; 35:e23881. [PMID: 34240756 PMCID: PMC8373329 DOI: 10.1002/jcla.23881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/09/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background Acute kidney injury (AKI) was characterized by loss of renal function, associated with chronic kidney disease, end‐stage renal disease, and length of hospital stay. Long non‐coding RNAs (lncRNAs) participated in AKI development and progression. Here, we aimed to investigate the roles and mechanisms of lncRNA MALAT1 in AKI. Methods AKI serum samples were obtained from 129 AKI patients. ROC analysis was conducted to confirm the diagnostic value of MALAT1 in differentiating AKI from healthy volunteers. After hypoxic treatment on HK‐2 cells, the expressions of inflammatory cytokines, MALAT1, miR‐204, APOL1, p65, and p‐p65, were measured by RT‐qPCR and Western blot assays. The targeted relationship between miR‐204 and MALAT1 or miR‐204 and APOL1 was determined by luciferase reporter assay and RNA pull‐down analysis. After transfection, CCK‐8, flow cytometry, and TUNEL staining assays were performed to evaluate the effects of MALAT1 and miR‐204 on AKI progression. Results From the results, lncRNA MALAT1 was strongly elevated in serum samples from AKI patients, with the high sensitivity and specificity concerning differentiating AKI patients from healthy controls. In vitro, we established the AKI cell model after hypoxic treatment. After experiencing hypoxia, we found significantly increased MALAT1, IL‐1β, IL‐6, and TNF‐α expressions along with decreased miR‐204 level. Moreover, the targeted relationship between MALAT1 and miR‐204 was confirmed. Silencing of MALAT1 could reverse hypoxia‐triggered promotion of HK‐2 cell apoptosis. Meanwhile, the increase of IL‐1β, IL‐6, and TNF‐α after hypoxia treatment could be repressed by MALAT1 knockdown as well. After co‐transfection with MALAT1 silencing and miR‐204 inhibition, we found that miR‐204 could counteract the effects of MALAT1 on HK‐2 cell progression and inflammation after under hypoxic conditions. Finally, NF‐κB signaling was inactivated while APOL1 expression was increased in HK‐2 cells after hypoxia treatment, and lncRNA MALAT1 inhibition reactivated NF‐κB signaling while suppressed APOL1 expression by sponging miR‐204. Conclusions Collectively, these results illustrated that knockdown of lncRNA MALAT1 could ameliorate AKI progression and inflammation by targeting miR‐204 through APOL1/NF‐κB signaling.
Collapse
Affiliation(s)
- Hai-Yuan Lu
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Guo-Yi Wang
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jin-Wen Zhao
- Department of Nephrology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hai-Tao Jiang
- Department of Orthopedics, Huai'an First People's Hospital, Huai'an, China
| |
Collapse
|
138
|
Albino AH, Zambom FFF, Foresto-Neto O, Oliveira KC, Ávila VF, Arias SCA, Seguro AC, Malheiros DMAC, Camara NOS, Fujihara CK, Zatz R. Renal Inflammation and Innate Immune Activation Underlie the Transition From Gentamicin-Induced Acute Kidney Injury to Renal Fibrosis. Front Physiol 2021; 12:606392. [PMID: 34305624 PMCID: PMC8293269 DOI: 10.3389/fphys.2021.606392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 06/11/2021] [Indexed: 01/15/2023] Open
Abstract
Subjects recovering from acute kidney injury (AKI) are at risk of developing chronic kidney disease (CKD). The mechanisms underlying this transition are unclear and may involve sustained activation of renal innate immunity, with resulting renal inflammation and fibrosis. We investigated whether the NF-κB system and/or the NLRP3 inflammasome pathway remain activated after the resolution of AKI induced by gentamicin (GT) treatment, thus favoring the development of CKD. Male Munich-Wistar rats received daily subcutaneous injections of GT, 80 mg/kg, for 9 days. Control rats received vehicle only (NC). Rats were studied at 1, 30, and 180 days after GT treatment was ceased. On Day 1, glomerular ischemia (ISCH), tubular necrosis, albuminuria, creatinine retention, and tubular dysfunction were noted, in association with prominent renal infiltration by macrophages and myofibroblasts, along with increased renal abundance of TLR4, IL-6, and IL1β. Regression of functional and structural changes occurred on Day 30. However, the renal content of IL-1β was still elevated at this time, while the local renin-angiotensin system remained activated, and interstitial fibrosis became evident. On Day 180, recurring albuminuria and mild glomerulosclerosis were seen, along with ISCH and unabated interstitial fibrosis, whereas macrophage infiltration was still evident. GT-induced AKI activates innate immunity and promotes renal inflammation. Persistence of these abnormalities provides a plausible explanation for the transition of AKI to CKD observed in a growing number of patients.
Collapse
Affiliation(s)
- Amanda Helen Albino
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Orestes Foresto-Neto
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Karin Carneiro Oliveira
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Victor Ferreira Ávila
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Simone Costa Alarcon Arias
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Antonio Carlos Seguro
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Niels Olsen Saraiva Camara
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil.,Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Clarice Kazue Fujihara
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Roberto Zatz
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
139
|
Portilla D, Xavier S. Role of intracellular complement activation in kidney fibrosis. Br J Pharmacol 2021; 178:2880-2891. [PMID: 33555070 DOI: 10.1111/bph.15408] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023] Open
Abstract
Increased expression of complement C1r, C1s and C3 in kidney cells plays an important role in the pathogenesis of kidney fibrosis. Our studies suggest that activation of complement in kidney cells with increased generation of C3 and its fragments occurs by activation of classical and alternative pathways. Single nuclei RNA sequencing studies in kidney tissue from unilateral ureteral obstruction mice show that increased synthesis of complement C3 and C5 occurs primarily in renal tubular epithelial cells (proximal and distal), while increased expression of complement receptors C3ar1 and C5ar1 occurs in interstitial cells including immune cells like monocytes/macrophages suggesting compartmentalization of complement components during kidney injury. Although global deletion of C3 and macrophage ablation prevent inflammation and reduced kidney tissue scarring, the development of mice with cell-specific deletion of complement components and their regulators could bring further insights into the mechanisms by which intracellular complement activation leads to fibrosis and progressive kidney disease. LINKED ARTICLES: This article is part of a themed issue on Canonical and non-canonical functions of the complement system in health and disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.14/issuetoc.
Collapse
Affiliation(s)
- Didier Portilla
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Sandhya Xavier
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
140
|
Marquez-Exposito L, Tejedor-Santamaria L, Santos-Sanchez L, Valentijn FA, Cantero-Navarro E, Rayego-Mateos S, Rodrigues-Diez RR, Tejera-Muñoz A, Marchant V, Sanz AB, Ortiz A, Goldschmeding R, Ruiz-Ortega M. Acute Kidney Injury is Aggravated in Aged Mice by the Exacerbation of Proinflammatory Processes. Front Pharmacol 2021; 12:662020. [PMID: 34239439 PMCID: PMC8258347 DOI: 10.3389/fphar.2021.662020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) is more frequent in elderly patients. Mechanisms contributing to AKI (tubular cell death, inflammatory cell infiltration, impaired mitochondrial function, and prolonged cell-cycle arrest) have been linked to cellular senescence, a process implicated in regeneration failure and progression to fibrosis. However, the molecular and pathological basis of the age-related increase in AKI incidence is not completely understood. To explore these mechanisms, experimental AKI was induced by folic acid (FA) administration in young (3-months-old) and old (1-year-old) mice, and kidneys were evaluated in the early phase of AKI, at 48 h. Tubular damage score, KIM-1 expression, the recruitment of infiltrating immune cells (mainly neutrophils and macrophages) and proinflammatory gene expression were higher in AKI kidneys of old than of young mice. Tubular cell death in FA-AKI involves several pathways, such as regulated necrosis and apoptosis. Ferroptosis and necroptosis cell-death pathways were upregulated in old AKI kidneys. In contrast, caspase-3 activation was only found in young but not in old mice. Moreover, the antiapoptotic factor BCL-xL was significantly overexpressed in old, injured kidneys, suggesting an age-related apoptosis suppression. AKI kidneys displayed evidence of cellular senescence, such as increased levels of cyclin dependent kinase inhibitors p16ink4a and p21cip1, and of the DNA damage response marker γH2AX. Furthermore, p21cip1 mRNA expression and nuclear staining for p21cip1 and γH2AX were higher in old than in young FA-AKI mice, as well as the expression of senescence-associated secretory phenotype (SASP) components (Il-6, Tgfb1, Ctgf, and Serpine1). Interestingly, some infiltrating immune cells were p21 or γH2AX positive, suggesting that molecular senescence in the immune cells (“immunosenescence”) are involved in the increased severity of AKI in old mice. In contrast, expression of renal protective factors was dramatically downregulated in old AKI mice, including the antiaging factor Klotho and the mitochondrial biogenesis driver PGC-1α. In conclusion, aging resulted in more severe AKI after the exposure to toxic compounds. This increased toxicity may be related to magnification of proinflammatory-related pathways in older mice, including a switch to a proinflammatory cell death (necroptosis) instead of apoptosis, and overactivation of cellular senescence of resident renal cells and infiltrating inflammatory cells.
Collapse
Affiliation(s)
- Laura Marquez-Exposito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Lucia Tejedor-Santamaria
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Laura Santos-Sanchez
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Elena Cantero-Navarro
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Raul R Rodrigues-Diez
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Antonio Tejera-Muñoz
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Ana B Sanz
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| |
Collapse
|
141
|
Deng X, Zhou C, Liao R, Guo Y, Wang Y, Li G, Wu J, Xu H, Hu Z, Pei G, Liao W, Yao Y, Yang Q, Zeng R, Xu G. Separated parabiont reveals the fate and lifespan of peripheral-derived immune cells in normal and ischaemia-induced injured kidneys. Open Biol 2021; 11:200340. [PMID: 34102079 PMCID: PMC8187026 DOI: 10.1098/rsob.200340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Immune cell infiltration plays a key role in acute kidney injury (AKI) to chronic kidney disease (CKD) progression. T lymphocytes, neutrophils, monocytes/macrophages and other immune cells regulate inflammation, tissue remodelling and repair. To determine the kinetics of accumulation of various immune cell populations, we established an animal model combining parabiosis and separation surgery to explore the fate and lifespan of peripheral leucocytes that migrate to the kidney. We found that peripheral T lymphocytes could survive for a long time (more than 14 days), whereas peripheral neutrophils survived for a short time in both healthy and ischaemia-induced damaged kidneys. Nearly half of the peripheral-derived macrophages disappeared after 14 days in normal kidneys, while their existing time in the inflammatory kidneys was prolonged. A fraction of F4/80high macrophages were renewed from the circulating monocyte pool. In addition, we found that after renal ischaemia reperfusion, neutrophils increased significantly in the early phase, and T lymphocytes mainly accumulated in the late stage, whereas macrophages infiltrated throughout AKI-CKD progression and were sustained longer in injured as opposed to normal kidneys. In conclusion, peripheral-derived macrophages, T lymphocytes and neutrophils exhibit different lifespans in the kidney, which may play different roles during AKI-CKD progression.
Collapse
Affiliation(s)
- Xuan Deng
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Cheng Zhou
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Ruichun Liao
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Yi Guo
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Yuxi Wang
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Guoli Li
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Jianliang Wu
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Huzi Xu
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Zhizhi Hu
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Guangchang Pei
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Wenhui Liao
- Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Ying Yao
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Qian Yang
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Rui Zeng
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| | - Gang Xu
- Division of Nephrology, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei 430030, People's Republic of China
| |
Collapse
|
142
|
Wan J, Zou G, He B, Zhang C, Zhu Y, Yin L, Lu Z. Development and External Validation a Novel Inflammation-Based Score for Acute Kidney Injury and Prognosis in Intensive Care Unit Patients. Int J Gen Med 2021; 14:2215-2226. [PMID: 34103975 PMCID: PMC8180284 DOI: 10.2147/ijgm.s311021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose We aimed to evaluate the predictive ability of an integrated score based on several inflammatory indices of acute kidney injury (AKI) in patients in the intensive care unit (ICU). Patients and Methods In this observational study, 2555 patients from the Medical Information Mart for Intensive Care III database were randomly assigned to the test set (n=1599) and internal validation set (n=656). Moreover, 412 coronary care unit patients from Zhongnan Hospital, Wuhan University were also included in the external validation set. The AKI-specific inflammatory index (ASII) was created using various inflammatory indices significantly associated with AKI. We further developed and validated two nomograms based on the ASII and other informative clinical features of AKI and prognosis. Results The ASII was calculated as 2.317×MLR+0.417×GPS+0.007×ALRI. In the training set, patients with a high ASII had a higher risk of incident AKI (odds ratio [OR], 5.33; 95% confidence index [CI], 3.60–7.88; P<0.001) than those with a low ASII with or without pre-existing chronic kidney disease. The nomograms for AKI and prognosis based on the ASII and other significant clinical characteristics had high predictive value in the prediction of AKI and prognosis in patients in the ICU. Moreover, the results in the internal validation set and in the external validation cohort were almost consistent with those in the training set. Conclusion The ASII is an AKI-specific tool based on the combination of available inflammatory indices. A high ASII is a strong predictor of a higher risk of AKI and worse survival outcomes in patients in the ICU.
Collapse
Affiliation(s)
- Jingjing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Gaorui Zou
- Department of Anesthesiology, Wuhan No. 1 Hospital, Wuhan, 430022, People's Republic of China
| | - Bo He
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Chao Zhang
- Department of Cardiology Electrocardiogram, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Yanfang Zhu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Lan Yin
- Department of Cardiology Electrocardiogram, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| |
Collapse
|
143
|
Arai H, Sato Y, Yanagita M. Fibroblast heterogeneity and tertiary lymphoid tissues in the kidney. Immunol Rev 2021; 302:196-210. [PMID: 33951198 PMCID: PMC8360208 DOI: 10.1111/imr.12969] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
Fibroblasts reside in various organs and support tissue structure and homeostasis under physiological conditions. Phenotypic alterations of fibroblasts underlie the development of diverse pathological conditions, including organ fibrosis. Recent advances in single‐cell biology have revealed that fibroblasts comprise heterogeneous subpopulations with distinct phenotypes, which exert both beneficial and detrimental effects on the host organs in a context‐dependent manner. In the kidney, phenotypic alterations of resident fibroblasts provoke common pathological conditions of chronic kidney disease (CKD), such as renal anemia and peritubular capillary loss. Additionally, in aged injured kidneys, fibroblasts provide functional and structural supports for tertiary lymphoid tissues (TLTs), which serve as the ectopic site of acquired immune reactions in various clinical contexts. TLTs are closely associated with aging and CKD progression, and the developmental stages of TLTs reflect the severity of renal injury. In this review, we describe the current understanding of fibroblast heterogeneity both under physiological and pathological conditions, with special emphasis on fibroblast contribution to TLT formation in the kidney. Dissecting the heterogeneous characteristics of fibroblasts will provide a promising therapeutic option for fibroblast‐related pathological conditions, including TLT formation.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Sato
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Medical Innovation Center, TMK Project, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
144
|
Abstract
Acute kidney injury (AKI), defined as a rapid decrease in glomerular filtration rate, is a common and devastating pathologic condition. AKI is associated with significant morbidity and subsequent chronic kidney disease (CKD) development. Regardless of the initial insult, CKD progression after AKI involves multiple types of cells, including proximal tubular cells, fibroblasts, and immune cells. Although the mechanisms underlying this AKI to CKD progression have been investigated extensively over the past decade, therapeutic strategies still are lacking. One of the reasons for this stems from the fact that AKI and its progression toward CKD is multifactorial and variable because it is dependent on patient background. In this review, we describe the current understanding of AKI and its maladaptive repair with a focus on proximal tubules and resident fibroblasts. Subsequently, we discuss the unique pathophysiology of AKI in the elderly, highlighting our recent finding of age-dependent tertiary lymphoid tissues.
Collapse
Affiliation(s)
- Yuki Sato
- Medical Innovation Center, TMK Project, Kyoto University, Kyoto, Japan; Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Takahashi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan.
| |
Collapse
|
145
|
Diptyanusa A, Phumratanaprapin W. Predictors and Outcomes of Dengue-Associated Acute Kidney Injury. Am J Trop Med Hyg 2021; 105:24-30. [PMID: 33939642 PMCID: PMC8274771 DOI: 10.4269/ajtmh.21-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Dengue viral infections present with a wide clinical spectrum ranging from asymptomatic to severe manifestations with organ involvement. The term "expanded dengue syndrome" has been commonly used to illustrate the unusual or atypical manifestations; acute kidney injury (AKI) is one of the atypical manifestations of this syndrome. The use of heterogeneous criteria to determine the presence of AKI in dengue patients due to the vast diversity in populations led to difficulties in assessing the true incidence of dengue-associated AKI. This review presents a variable, but often high, frequency of dengue-associated AKI among vastly diverse populations with various disease severities. Dengue-associated AKI is not an uncommon complication, and its importance has often been neglected during the management of dengue patients. The risk factors and certain clinical and laboratory findings commonly reported among dengue patients with AKI should be considered to support a timely diagnosis and case management. This review highlights the need for clinicians to be aware of dengue-associated AKI to reduce the morbidity and mortality associated with this common and important tropical disease.
Collapse
Affiliation(s)
- Ajib Diptyanusa
- Center for Tropical Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Weerapong Phumratanaprapin
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
146
|
Xia J, Cao W. Epigenetic modifications of Klotho expression in kidney diseases. J Mol Med (Berl) 2021; 99:581-592. [PMID: 33547909 DOI: 10.1007/s00109-021-02044-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/10/2020] [Accepted: 01/20/2021] [Indexed: 12/21/2022]
Abstract
Developments of many renal diseases are substantially influenced by epigenetic modifications of numerous genes, mainly mediated by DNA methylations, histone modifications, and microRNA interference; however, not all gene modifications causally affect the disease onset or progression. Klotho is a critical gene whose repressions in various pathological conditions reportedly involve epigenetic regulatory mechanisms. Klotho is almost unexceptionally repressed early after acute or chronic renal injuries and its levels inversely correlated with the disease progression and severity. Moreover, the strategies of Klotho derepression via epigenetic modulations beneficially change the pathological courses both in vitro and in vivo. Hence, Klotho is not only considered a biomarker of the renal disease but also a potential or even an ideal target of therapeutic epigenetic intervention. Here, we summarize and discuss studies that investigate the Klotho repression and intervention in renal diseases from an epigenetic point of view. These information might shed new sights into the effective therapeutic strategies to prevent and treat various renal disorders.
Collapse
Affiliation(s)
- Jinkun Xia
- Center for Organ Fibrosis and Remodeling Research, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China
| | - Wangsen Cao
- Center for Organ Fibrosis and Remodeling Research, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China.
| |
Collapse
|
147
|
Jeon J, Lee K, Yang KE, Lee JE, Kwon GY, Huh W, Kim DJ, Kim YG, Jang HR. Dietary Modification Alters the Intrarenal Immunologic Micromilieu and Susceptibility to Ischemic Acute Kidney Injury. Front Immunol 2021; 12:621176. [PMID: 33777001 PMCID: PMC7991094 DOI: 10.3389/fimmu.2021.621176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022] Open
Abstract
The versatility of the intrarenal immunologic micromilieu through dietary modification and the subsequent effects on susceptibility to ischemic acute kidney injury (AKI) are unclear. We investigated the effects of high-salt (HS) or high-fat (HF) diet on intrarenal immunologic micromilieu and development of ischemic AKI using murine ischemic AKI and human kidney-2 (HK-2) cell hypoxia models. Four different diet regimens [control, HF, HS, and high-fat diet with high-salt (HF+HS)] were provided individually to groups of 9-week-old male C57BL/6 mice for 1 or 6 weeks. After a bilateral ischemia-reperfusion injury (BIRI) operation, mice were sacrificed on day 2 and renal injury was assessed with intrarenal leukocyte infiltration. Human kidney-2 cells were treated with NaCl or lipids. The HF diet increased body weight and total cholesterol, whereas the HF+HS did not. Although the HF or HS diet did not change total leukocyte infiltration at 6 weeks, the HF diet and HF+HS diet increased intrarenal CD8 T cells. Plasma cells increased in the HF and HS diet groups. The expression of proinflammatory cytokines including TNF-α, IFN-γ, MCP-1, and RANTES was increased by the HF or HS diet, and intrarenal VEGF decreased in the HS and HF+HS diet groups at 6 weeks. Deterioration of renal function following BIRI tended to be aggravated by the HF or HS diet. High NaCl concentration suppressed proliferation and enhanced expression of TLR-2 in hypoxic HK-2 cells. The HF or HS diet can enhance susceptibility to ischemic AKI by inducing proinflammatory changes to the intrarenal immunologic micromilieu.
Collapse
Affiliation(s)
- Junseok Jeon
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyeong Eun Yang
- Division of Scientific Instrumentation and Management, Korea Basic Science Institute, Daejeon, South Korea
| | - Jung Eun Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ghee Young Kwon
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Wooseong Huh
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Dae Joong Kim
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yoon-Goo Kim
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
148
|
Tanaka M, Saka-Tanaka M, Ochi K, Fujieda K, Sugiura Y, Miyamoto T, Kohda H, Ito A, Miyazawa T, Matsumoto A, Aoe S, Miyamoto Y, Tsuboi N, Maruyama S, Suematsu M, Yamasaki S, Ogawa Y, Suganami T. C-type lectin Mincle mediates cell death-triggered inflammation in acute kidney injury. J Exp Med 2021; 217:152022. [PMID: 32797195 PMCID: PMC7596812 DOI: 10.1084/jem.20192230] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/08/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence indicates that cell death triggers sterile inflammation and that impaired clearance of dead cells causes nonresolving inflammation; however, the underlying mechanisms are still unclear. Here, we show that macrophage-inducible C-type lectin (Mincle) senses renal tubular cell death to induce sustained inflammation after acute kidney injury in mice. Mincle-deficient mice were protected against tissue damage and subsequent atrophy of the kidney after ischemia–reperfusion injury. Using lipophilic extract from the injured kidney, we identified β-glucosylceramide as an endogenous Mincle ligand. Notably, free cholesterol markedly enhanced the agonistic effect of β-glucosylceramide on Mincle. Moreover, β-glucosylceramide and free cholesterol accumulated in dead renal tubules in proximity to Mincle-expressing macrophages, where Mincle was supposed to inhibit clearance of dead cells and increase proinflammatory cytokine production. This study demonstrates that β-glucosylceramide in combination with free cholesterol acts on Mincle as an endogenous ligand to induce cell death–triggered, sustained inflammation after acute kidney injury.
Collapse
Affiliation(s)
- Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Marie Saka-Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozue Ochi
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kumiko Fujieda
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Tomofumi Miyamoto
- Department of Natural Products Chemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiro Kohda
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ayaka Ito
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taiki Miyazawa
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akira Matsumoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seiichiro Aoe
- Department of Home Economics, Otsuma Women's University, Tokyo, Japan
| | - Yoshihiro Miyamoto
- Division of Genomic Diagnosis and Healthcare, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Naotake Tsuboi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology, Tokyo, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
149
|
Pai MP. Antimicrobial Dosing in Specific Populations and Novel Clinical Methodologies: Kidney Function. Clin Pharmacol Ther 2021; 109:952-957. [PMID: 33523498 DOI: 10.1002/cpt.2179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022]
Abstract
Kidney function is a common parameter used to define antimicrobial drug dosage and frequency of administration. Several methods exist to measure kidney function but for pragmatic reasons rely on estimated kidney function equations based on the endogenous biomarker serum creatinine and common clinical variables. Current regulatory guidance on the design of studies in patients with abnormal kidney function in the United States also recommend consideration of estimated kidney function for this reason. Over the past few decades, alternate endogenous biomarkers, administration of exogenous biomarkers for noninvasive measurement, use of probe substrates to characterize individual kidney drug clearance pathways, modifications to conventional equations to account for time-varying clearance, and improved clinical trial modeling and simulation to factor in these uncertainties have occurred. Furthermore, major changes to kidney replacement therapy delivery in the outpatient, inpatient, and at-home setting are occurring. Antimicrobial drug dose adjustment in this diverse population is complex and in a state of flux due to technical innovations. Over-reliance on kidney function estimates to guide drug dosing in patients with infectious diseases can bias underdosing especially among the acutely ill. A holistic approach to drug dose adjustment in patients with abnormal kidney function is necessary to optimize clinical outcomes.
Collapse
Affiliation(s)
- Manjunath P Pai
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
150
|
McWilliam SJ, Wright RD, Welsh GI, Tuffin J, Budge KL, Swan L, Wilm T, Martinas IR, Littlewood J, Oni L. The complex interplay between kidney injury and inflammation. Clin Kidney J 2021; 14:780-788. [PMID: 33777361 PMCID: PMC7986351 DOI: 10.1093/ckj/sfaa164] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) has gained significant attention following patient safety alerts about the increased risk of harm to patients, including increased mortality and hospitalization. Common causes of AKI include hypovolaemia, nephrotoxic medications, ischaemia and acute glomerulonephritis, although in reality it may be undetermined or multifactorial. A period of inflammation either as a contributor to the kidney injury or resulting from the injury is almost universally seen. This article was compiled following a workshop exploring the interplay between injury and inflammation. AKI is characterized by some degree of renal cell death through either apoptosis or necrosis, together with a strong inflammatory response. Studies interrogating the resolution of renal inflammation identify a whole range of molecules that are upregulated and confirm that the kidneys are able to intrinsically regenerate after an episode of AKI, provided the threshold of damage is not too high. Kidneys are unable to generate new nephrons, and dysfunctional or repeated episodes will lead to further nephron loss that is ultimately associated with the development of renal fibrosis and chronic kidney disease (CKD). The AKI to CKD transition is a complex process mainly facilitated by maladaptive repair mechanisms. Early biomarkers mapping out this process would allow a personalized approach to identifying patients with AKI who are at high risk of developing fibrosis and subsequent CKD. This review article highlights this process and explains how laboratory models of renal inflammation and injury assist with understanding the underlying disease process and allow interrogation of medications aimed at targeting the mechanistic interplay.
Collapse
Affiliation(s)
- Stephen J McWilliam
- Department of Paediatric Pharmacology, Alder Hey Children’s Hospital, Liverpool, UK
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Rachael D Wright
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jack Tuffin
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kelly L Budge
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Laura Swan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Thomas Wilm
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Ioana-Roxana Martinas
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - James Littlewood
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Nephrology, Royal Liverpool University Hospital, Liverpool, UK
| | - Louise Oni
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Paediatric Nephrology, Alder Hey Children’s NHS Foundation Trust Hospital, Liverpool, UK
| |
Collapse
|