1
|
Kong FX, Liu H, Xu T, Li SJ, Li W, Lu H, Ma NN, Wang YL, Shi JH, Yang YR, Wang FL. RG108 attenuates acute kidney injury by inhibiting P38 MAPK/FOS and JNK/JUN pathways. Int Immunopharmacol 2024; 142:113077. [PMID: 39265353 DOI: 10.1016/j.intimp.2024.113077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/16/2024] [Accepted: 09/01/2024] [Indexed: 09/14/2024]
Abstract
Acute kidney injury (AKI) is an important clinical syndrome characterised by a sudden decline in renal function, often accompanied by renal inflammation and tubular epithelial cell damage. It has been reported that inhibiting DNA methylation significantly suppress the progression of AKI. In the current study, we investigate the effect of the DNA methyltransferase (DNMT) inhibitor RG108 in cisplatin- and hypoxia-reoxygenation-induced AKI. The expression of kidney injury molecules and inflammatory factors was examined by immunofluorescence, Western blotting and Real-time PCR. The results demonstrated that RG108 treatment significantly reduced kidney inflammation and injury. Furthermore, RNA-seq analysis was performed to reveal the regulatory mechanism of RG108 in AKI. The expression of the FOS and JUN genes, which are downstream of the MAPK pathway, were significant increased in AKI. Meanwhile, the expression of FOS and JUN were both inhibited by RG108, which is similar to what we found treatment with a specific JNK inhibitor and a specific p38 MAPK inhibitor, and thus attenuated renal inflammation and injury. In conclusion, we suggest that RG108 inhibits P38 MAPK/FOS and JNK/JUN pathways and attenuates renal injury and inflammatory responses. In these results, RG108 may become a novel MAPK pathway inhibitor and a clinical candidate for the treatment of AKI.
Collapse
Affiliation(s)
- Fan-Xu Kong
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Second People's Hospital of Hefei, Hefei 230011, Anhui, China
| | - Hui Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Tao Xu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Second People's Hospital of Hefei, Hefei 230011, Anhui, China
| | - Shuang-Jian Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Hao Lu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Nan-Nan Ma
- Jiangsu Huaiyin Maternity and Children's Hospital, Jiangsu 223399, China
| | - Yun-Long Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ji-Hong Shi
- Jiangsu Huaiyin Maternity and Children's Hospital, Jiangsu 223399, China
| | - Ya-Ru Yang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Feng-Ling Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; The Second People's Hospital of Hefei, Hefei 230011, Anhui, China.
| |
Collapse
|
2
|
Pereira DP, Moreira BS, Rodrigues MA, Magalhães LF, Branco LDO, Reis NS, Borin-Crivellenti S, Crivellenti LZ. Renal protective potential of pentoxifylline, chlorpromazine, and lovastatin in ischemia-reperfusion injury: An experimental study. PLoS One 2024; 19:e0308649. [PMID: 39413126 PMCID: PMC11482703 DOI: 10.1371/journal.pone.0308649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/27/2024] [Indexed: 10/18/2024] Open
Abstract
This study aimed to evaluate the ability of pentoxifylline when compared to lovastatin and chlorpromazine as nephroprotective substances in cases of renal ischemia and reperfusion syndrome (IRI). A total of 36 adult male animals were randomly allocated into four groups (untreated control group, pentoxifylline group, lovastatin group, and chlorpromazine group), each consisting of nine animals. All groups were submitted to experimental ischemia and reperfusion procedures. The animals were evaluated 24, 72 and 120 hours after IRI, including physical examinations, serum urea and creatinine measurements, as well as histopathological, morphometric, and stereological analyses of the renal tissue. Results indicated that 24 hours after IRI, only chlorpromazine was effective in controlling azotemia. At the 72-hour mark, both chlorpromazine and pentoxifylline exhibited efficacy. After 120 hours, all three substances demonstrated renal protective qualities. Pentoxifylline was the most effective in preserving the structural integrity of kidney tissue, followed by chlorpromazine. In conclusion, all three treatments (pentoxifylline, chlorpromazine, and lovastatin) were effective. Pentoxifylline proved to be promising in the response against acute tubular necrosis, although chlorpromazine presented earlier renoprotective effects in terms of maintaining renal function.
Collapse
Affiliation(s)
- Daniel Peixoto Pereira
- Veterinary Teaching Hospital/ Animal Science Graduate Program, Franca University (UNIFRAN), Franca, São Paulo, Brazil
| | - Brunna Silva Moreira
- Graduate Program in Veterinary Science, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Marcela Aldrovani Rodrigues
- Veterinary Teaching Hospital/ Animal Science Graduate Program, Franca University (UNIFRAN), Franca, São Paulo, Brazil
| | - Larissa Fernandes Magalhães
- Veterinary Teaching Hospital/ Animal Science Graduate Program, Franca University (UNIFRAN), Franca, São Paulo, Brazil
| | - Luana de Oliveira Branco
- Graduate Program in Veterinary Science, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Natani Silva Reis
- Graduate Program in Veterinary Science, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Sofia Borin-Crivellenti
- Graduate Program in Veterinary Science, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | | |
Collapse
|
3
|
Kanai M, Nishino T, Daassi D, Kimura A, Liao CW, Javanfekr Shahri Z, Wakimoto A, Gogoleva N, Usui T, Morito N, Arita M, Takahashi S, Hamada M. MAFB in Macrophages Regulates Prostaglandin E2-Mediated Lipid Mediator Class Switch through ALOX15 in Ischemic Acute Kidney Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1212-1224. [PMID: 39230290 PMCID: PMC11457724 DOI: 10.4049/jimmunol.2300844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 07/31/2024] [Indexed: 09/05/2024]
Abstract
Monocytes and macrophages express the transcription factor MAFB (V-maf musculoaponeurotic fibrosarcoma oncogene homolog B) and protect against ischemic acute kidney injury (AKI). However, the mechanism through which MAFB alleviates AKI in macrophages remains unclear. In this study, we induced AKI in macrophage lineage-specific Mafb-deficient mice (C57BL/6J) using the ischemia-reperfusion injury model to analyze these mechanisms. Our results showed that MAFB regulates the expression of Alox15 (arachidonate 15-lipoxygenase) in macrophages during ischemic AKI. The expression of ALOX15 was significantly decreased at the mRNA and protein levels in macrophages that infiltrated the kidneys of macrophage-specific Mafb-deficient mice at 24 h after ischemia-reperfusion injury. ALOX15 promotes the resolution of inflammation under acute conditions by producing specialized proresolving mediators by oxidizing essential fatty acids. Therefore, MAFB in macrophages promotes the resolution of inflammation in ischemic AKI by regulating the expression of Alox15. Moreover, MAFB expression in macrophages is upregulated via the COX-2/PGE2/EP4 pathway in ischemic AKI. Our in vitro assay showed that MAFB regulates the expression of Alox15 under the COX-2/PGE2/EP4 pathway in macrophages. PGE2 mediates the lipid mediator (LM) class switch from inflammatory LMs to specialized proresolving mediators. Therefore, MAFB plays a key role in the PGE2-mediated LM class switch by regulating the expression of Alox15. Our study identified a previously unknown mechanism by which MAFB in macrophages alleviates ischemic AKI and provides new insights into regulating the LM class switch in acute inflammatory conditions.
Collapse
Affiliation(s)
- Maho Kanai
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Teppei Nishino
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Tsukuba Medical Center Hospital, Tsukuba, Japan
| | - Dhouha Daassi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akari Kimura
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Ching-Wei Liao
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Zeynab Javanfekr Shahri
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Arata Wakimoto
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Natalia Gogoleva
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Toshiaki Usui
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoki Morito
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Science, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan; and
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
4
|
Lin WY, Cheng YH, Liu PY, Hsu SP, Lin SC, Chien CT. Carvedilol through ß1-Adrenoceptor blockade ameliorates glomerulonephritis via inhibition of oxidative stress, apoptosis, autophagy, ferroptosis, endoplasmic reticulum stress and inflammation. Biochem Pharmacol 2024; 230:116570. [PMID: 39401703 DOI: 10.1016/j.bcp.2024.116570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/28/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024]
Abstract
Glomerulonephritis (GN) is one of the main causes of end stage renal disease and requires an effective treatment for inhibiting GN. Renal nerves through efferent (RENA) and afferent (RANA) innervation to glomeruli regulate the glomerular function. We delineated the role of RENA and RANA on anti-Thy1.1-induced GN. Female Wistar rats were divided into Control, Thy1.1 plus anti-Thy1.1, bilaterally renal nerve denervation (DNX) plus anti-Thy1.1, and topical capsaicin to bilateral renal nerves for selective ablation of RANA (DNAX) plus anti-Thy1.1. We examined RANA and RENA response to anti-Thy1.1 and compared the effect of DNX or DNAX on urinary oxidative stress, renal gp91, tyrosine hydroxylase (TH), calcitonin gene-related peptide (CGRP), apoptosis, autophagy, ferroptosis, antioxidant enzymes, endoplasmic reticulum (ER) stress and inflammation by western blot. Anti-Thy1.1 significantly enhanced RENA, but did not affect RANA. DNX significantly decreased TH and CGRP expression, whereas DNAX only reduced CGRP expression. Anti-Thy1.1 significantly increased glomerulosclerosis injury, urinary protein, electron paramagnetic resonance signals of alpha-(4-pyridyl-N-oxide)-N-tert-butylnitrone adducts, 8-isoprostane and nitrotyrosine levels, NADPH oxidase gp91phox (gp91), macrophage/monocyte (ED-1), GRP-78, Beclin-1/LC3-II, Bax/caspase-3/poly(ADP-ribose) polymerase expression, inflammatory cytokines levels and decreased renal Copper/Zinc superoxide dismutase, Cystine/glutamate transporter (xCT) and Glutathione peroxidase 4 (GPX4) expression vs. Control. The enhanced oxidative parameters or reduced antioxidant defense by anti-Thy1.1 were significantly attenuated by DNX but not DNAX. Additionally, oral ß1-adrenoceptor antagonist-Carvedilol at an early stage reduced anti-Thy1.1 increased proteinuria level and oxidative parameters. Our data suggest that DNX and ß1-adrenoceptor antagonist-Carvedilol efficiently attenuate oxidative stress, inflammation, ER stress, autophagy, ferroptosis and apoptosis in GN.
Collapse
Affiliation(s)
- Wei-Yu Lin
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan; Department of Urology, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 24213, Taiwan
| | - Yu-Hsuan Cheng
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Pei-Yu Liu
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Shih-Ping Hsu
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan; Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan; Department of Industrial Management, Oriental Institute of Technology, New Taipei City 220, Taiwan; General Education Center, Lunghwa University of Science and Technology, Taoyuan, Taiwan.
| | - San-Chi Lin
- Division of Renal Section, Department of Internal Medicine, Keelung Hospital, Ministry of Health and Welfare, Keelung City 201, Taiwan.
| | - Chiang-Ting Chien
- Department of Life Science, School of Life Science, College of Science, National Taiwan Normal University, Taipei 11677, Taiwan.
| |
Collapse
|
5
|
Wei Q, Huang J, Livingston MJ, Wang S, Dong G, Xu H, Zhou J, Dong Z. Pseudogene GSTM3P1 derived long non-coding RNA promotes ischemic acute kidney injury by target directed microRNA degradation of kidney-protective mir-668. Kidney Int 2024; 106:640-657. [PMID: 39074555 PMCID: PMC11416318 DOI: 10.1016/j.kint.2024.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/31/2024]
Abstract
Long non-coding RNAs (lncRNAs) are a group of epigenetic regulators that have been implicated in kidney diseases including acute kidney injury (AKI). However, very little is known about the specific lncRNAs involved in AKI and the mechanisms underlying their pathologic roles. Here, we report a new lncRNA derived from the pseudogene GSTM3P1, which mediates ischemic AKI by interacting with and promoting the degradation of mir-668, a kidney-protective microRNA. GSTM3P1 and its mouse orthologue Gstm2-ps1 were induced by hypoxia in cultured kidney proximal tubular cells. In mouse kidneys, Gstm2-ps1 was significantly upregulated in proximal tubules at an early stage of ischemic AKI. This transient induction of Gstm2-ps1 depends on G3BP1, a key component in stress granules. GSTM3P1 overexpression increased kidney proximal tubular apoptosis after ATP depletion, which was rescued by mir-668. Notably, kidney proximal tubule-specific knockout of Gstm2-ps1 protected mice from ischemic AKI, as evidenced by improved kidney function, diminished tubular damage and apoptosis, and reduced kidney injury biomarker (NGAL) induction. To test the therapeutic potential, Gstm2-ps1 siRNAs were introduced into cultured mouse proximal tubular cells or administered to mice. In cultured cells, Gstm2-ps1 knockdown suppressed ATP depletion-associated apoptosis. In mice, Gstm2-ps1 knockdown ameliorated ischemic AKI. Mechanistically, both GSTM3P1 and Gstm2-ps1 possessed mir-668 binding sites and downregulated the mature form of mir-668. Specifically, GSTM3P1 directly bound to mature mir-668 to induce its decay via target-directed microRNA degradation. Thus, our results identify GSTM3P1 as a novel lncRNA that promotes kidney tubular cell death in AKI by binding mir-668 to inducing its degradation.
Collapse
Affiliation(s)
- Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.
| | - Jing Huang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Man Jiang Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Shixuan Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hongyan Xu
- Department of Biostatistics, Data Science and Epidemiology, School of Public Health, Augusta University, Augusta, Georgia, USA
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Charlie Norwood VA Medical Center, Augusta, Georgia, USA.
| |
Collapse
|
6
|
Luo Z, Li Y, Xie M, Yi S, Xu S, Luo J. Soluble suppression of tumorigenicity 2 associated with contrast-induced acute kidney injury in patients with STEMI. Int Urol Nephrol 2024:10.1007/s11255-024-04204-4. [PMID: 39264493 DOI: 10.1007/s11255-024-04204-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Contrast-induced acute kidney injury (CI-AKI) is a common complication after percutaneous coronary intervention (PCI) in patients with ST-segment elevation myocardial infarction (STEMI). Soluble suppression of tumorigenicity 2 (sST2) is associated with AKI. However, the relationship between sST2 and CI-AKI is unclear. This study aimed to investigate the relationship between sST2 and CI-AKI in patients with STEMI. METHODS This was a single-center retrospective observational study. Patients diagnosed with STEMI in the Yichun People's Hospital from February 2020 to May 2024 were continuously included. CI-AKI was defined as an increase in serum creatinine of at least 50% or 0.3 mg/dL from baseline within 48-72 h after contrast exposure. RESULTS The incidence of CI-AKI after PCI was 12.4% (98/791). Univariate analysis showed that age, fasting plasma glucose, diabetes mellitus, Killip class, left ventricular ejection fraction, estimated glomerular filtration rate, high sensitivity troponin T, N-terminal pro-B-type natriuretic peptide, and sST2 were associated with CI-AKI. The above factors were included in a multivariate analysis, which showed that sST2 was an independent factor for CI-AKI after PCI. The restricted cubic splines showed a nonlinear dose-response relationship between sST2 and CI-AKI (P < 0.001). The integration of the sST2 could significantly improve the ability of the model to identify CI-AKI (NRI 0.681, 95% CI 0.474-0.887; IDI 0.063, 95% CI 0.038-0.099). CONCLUSION Elevated sST2 is an independent risk factor for CI-AKI after PCI in patients with STEMI. Integration of sST2 can significantly improve the risk model for CI-AKI.
Collapse
Affiliation(s)
- Ziyun Luo
- Department of Nephrology, Yichun People's Hospital, Yichun, 336000, Jiangxi, China
| | - Yong Li
- Department of Cardiology, The First People's Hospital of Yuhang District, Hangzhou, 311100, Zhejiang, China
| | - Minjuan Xie
- Department of Medicine, Yichun University, Yichun, 336000, Jiangxi, China
| | - Song Yi
- Department of Cardiology, Yichun People's Hospital, Yichun, 336000, Jiangxi, China
| | - Shizhang Xu
- Department of Nephrology, Yichun People's Hospital, Yichun, 336000, Jiangxi, China
| | - Jun Luo
- Department of Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
7
|
Border S, Ferreira RM, Lucarelli N, Manthey D, Kumar S, Paul A, Mimar S, Naglah A, Cheng YH, Barisoni L, Ray J, Strekalova Y, Rosenberg AZ, Tomaszewski JE, Hodgin JB, El-Achkar TM, Jain S, Eadon MT, Sarder P. FUSION: A web-based application for in-depth exploration of multi-omics data with brightfield histology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602778. [PMID: 39026885 PMCID: PMC11257503 DOI: 10.1101/2024.07.09.602778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Spatial -OMICS technologies facilitate the interrogation of molecular profiles in the context of the underlying histopathology and tissue microenvironment. Paired analysis of histopathology and molecular data can provide pathologists with otherwise unobtainable insights into biological mechanisms. To connect the disparate molecular and histopathologic features into a single workspace, we developed FUSION (Functional Unit State IdentificatiON in WSIs [Whole Slide Images]), a web-based tool that provides users with a broad array of visualization and analytical tools including deep learning-based algorithms for in-depth interrogation of spatial -OMICS datasets and their associated high-resolution histology images. FUSION enables end-to-end analysis of functional tissue units (FTUs), automatically aggregating underlying molecular data to provide a histopathology-based medium for analyzing healthy and altered cell states and driving new discoveries using "pathomic" features. We demonstrate FUSION using 10x Visium spatial transcriptomics (ST) data from both formalin-fixed paraffin embedded (FFPE) and frozen prepared datasets consisting of healthy and diseased tissue. Through several use-cases, we demonstrate how users can identify spatial linkages between quantitative pathomics, qualitative image characteristics, and spatial --omics.
Collapse
Affiliation(s)
- Samuel Border
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | | | - Nicholas Lucarelli
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | | | - Suhas Kumar
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Anindya Paul
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Sayat Mimar
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Ahmed Naglah
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Ying-Hua Cheng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Laura Barisoni
- Department of Pathology, Division of AI and Computational Pathology, Duke University, Durham, NC
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC
| | - Jessica Ray
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL
| | - Yulia Strekalova
- College of Public Health and Health Professions, University of Florida, Gainesville, FL
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine Baltimore, MD
| | - John E Tomaszewski
- Department of Pathology & Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | | | - Tarek M El-Achkar
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Indianapolis VA Medical Center, Indianapolis, IN
| | - Sanjay Jain
- Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, MO
| | - Michael T Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Pinaki Sarder
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| |
Collapse
|
8
|
Ahmed MA, Kamel EO, Abd-Eldayem AM. Role of cAMP/pCREB and GSK-3β/NF-κB p65 signaling pathways in the renoprotective effect of mirabegron against renal ischemia-reperfusion injury in rats. Eur J Pharmacol 2024; 974:176617. [PMID: 38679120 DOI: 10.1016/j.ejphar.2024.176617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Acute kidney injury and other renal disorders are thought to be primarily caused by renal ischemia-reperfusion (RIR). Cyclic adenosine monophosphate (cAMP) has plenty of physiological pleiotropic effects and preserves tissue integrity and functions. This research aimed to examine the potential protective effects of the β3-adrenergic receptors agonist mirabegron in a rat model of RIR and its underlying mechanisms. Male rats enrolled in this work were given an oral dose of 30 mg/kg mirabegron for two days before surgical induction of RIR. Renal levels of kidney injury molecule-1 (KIM-1), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), Interleukin-10 (IL-10), cAMP, cAMP-responsive element binding protein (pCREB), and glycogen synthase kinase-3 beta (GSK-3β) were assessed along with blood urea nitrogen and serum creatinine. Additionally, caspase-3 and nuclear factor-kappa B (NF-κB) p65 were explored by immunohistochemical analysis. Renal specimens were inspected for histopathological changes. RIR led to renal tissue damage with elevated blood urea nitrogen and serum creatinine levels. The renal KIM-1, MCP-1, TNF-α, and GSK-3β were significantly increased, while IL-10, cAMP, and pCREB levels were reduced. Moreover, upregulation of caspase-3 and NF-κB p65 protein expression was seen in RIR rats. Mirabegron significantly reduced kidney dysfunction, histological abnormalities, inflammation, and apoptosis in the rat renal tissues. Mechanistically, mirabegron mediated these effects via modulation of cAMP/pCREB and GSK-3β/NF-κB p65 signaling pathways. Mirabegron administration could protect renal tissue and maintain renal function against RIR.
Collapse
Affiliation(s)
- Marwa A Ahmed
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Esam O Kamel
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Ahmed M Abd-Eldayem
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Pharmacology, Faculty of Medicine, Merit University, Sohag, Egypt.
| |
Collapse
|
9
|
Patel NS, Herzog I, Dunn C, Merchant AM. Impact of Operative Approach on Acute Kidney Injury Risk Prediction Models for Colectomy. J Surg Res 2024; 299:224-236. [PMID: 38776578 DOI: 10.1016/j.jss.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 04/07/2024] [Accepted: 04/21/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Acute kidney injury (AKI) is a serious postoperative complication associated with increased morbidity and mortality. Identifying patients at risk for AKI is important for risk stratification and management. This study aimed to develop an AKI risk prediction model for colectomy and determine if the operative approach (laparoscopic versus open) alters the influence of predictive factors through an interaction term analysis. METHODS The American College of Surgeons National Surgical Quality Improvement Program database was analyzed from 2005 to 2019. Patients undergoing laparoscopic and open colectomy were identified and propensity score matched. Multivariable logistic regression identified significant preoperative demographic, comorbidity, and laboratory value predictors of AKI. The predictive ability of a baseline model consisting of these variables was compared to a proposed model incorporating interaction terms between operative approach and predictor variables using the likelihood ratio test, c-statistic, and Brier score. Shapley Additive Explanations values assessed relative importance of significant predictors. RESULTS 252,372 patients were included in the analysis. Significant AKI predictors were hypertension, age, sex, race, body mass index, smoking, diabetes, preoperative sepsis, Congestive heart failure, preoperative creatinine, preoperative albumin, and operative approach (P < 0.001). The proposed model with interaction terms had improved predictive ability per the likelihood ratio test (P < 0.05) but had no statistically significant interaction terms. C-statistic and Brier scores did not improve. Shapley Additive Explanations analysis showed hypertension had the highest importance. The importance of age and diabetes showed some variation between operative approaches. CONCLUSIONS While the inclusion of interaction terms collectively improved AKI prediction, no individual operative approach interaction terms were significant. Including operative approach interactions may enhance predictive ability of AKI risk models for colectomy.
Collapse
Affiliation(s)
| | - Isabel Herzog
- Rutgers New Jersey Medical School, Newark, New Jersey
| | - Colin Dunn
- Department of Surgery, Good Samaritan Hospital, San Jose, California
| | - Aziz M Merchant
- Rutgers New Jersey Medical School, Newark, New Jersey; Division of General and Minimally Invasive Surgery, Department of Surgery, Hackensack Meridian School of Medicine, JFK Hackensack Meridian Medical Center, Edison, New Jersey.
| |
Collapse
|
10
|
Pula W, Ganugula R, Esposito E, Ravi Kumar MNV, Arora M. Engineered urolithin A-laden functional polymer-lipid hybrid nanoparticles prevent cisplatin-induced proximal tubular injury in vitro. Eur J Pharm Biopharm 2024; 200:114334. [PMID: 38768764 PMCID: PMC11262884 DOI: 10.1016/j.ejpb.2024.114334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Functional polymer-lipid hybrid nanoparticles (H-NPs) are a promising class of nanocarriers that combine the benefits of polymer and lipid nanoparticles, offering biocompatibility, structural stability, high loading capacity, and, most importantly, superior surface functionalization. Here, we report the synthesis and design of highly functional H-NPs with specificity toward the transferrin receptor (TfR), using a small molecule ligand, gambogic acid (GA). A fluorescence study revealed the molecular orientation of H-NPs, where the lipid-dense core is surrounded by a polymer exterior, functionalized with GA. Urolithin A, an immunomodulator and anti-inflammatory agent, served as a model drug-like compound to prepare H-NPs via traditional emulsion-based techniques, where H-NPs led to smaller particles (132 nm) and superior entrapment efficiencies (70 % at 10 % drug loading) compared to GA-conjugated polymeric nanoparticles (P-NPs) (157 nm and 52 % entrapment efficiency) and solid lipid nanoparticles (L-NPs) (186 nm and 29 % entrapment efficiency). H-NPs showed superior intracellular accumulation compared to individual NPs using human small intestinal epithelial (FHs 74) cells. The in vitro efficacy was demonstrated by flow cytometry analysis, in which UA-laden H-NPs showed excellent anti-inflammatory properties in cisplatin-induced injury in healthy human proximal tubular cell (HK2) model by decreasing the TLR4, NF-κβ, and IL-β expression. This preliminary work highlights the potential of H-NPs as a novel functional polymer-lipid drug delivery system, establishing the foundation for future research on its therapeutic potential in addressing chemotherapy-induced acute kidney injury in cancer patients.
Collapse
Affiliation(s)
- W Pula
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Fossato di Mortara, 19-44121 Ferrara, Italy; The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States
| | - R Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States; Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States; Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States; Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States
| | - E Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Fossato di Mortara, 19-44121 Ferrara, Italy
| | - M N V Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States; Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States; Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States; Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States; Chemical and Biological Engineering, University of Alabama, SEC 3448, Tuscaloosa, AL 35487, United States; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - M Arora
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States; Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States; Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States; Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States.
| |
Collapse
|
11
|
Kong Y, Chen X, Liu F, Tang J, Zhang Y, Zhang X, Zhang L, Zhang T, Wang Y, Su M, Zhang Q, Chen H, Zhou D, Yi F, Liu H, Fu Y. Ultrasmall Polyphenol-NAD + Nanoparticle-Mediated Renal Delivery for Mitochondrial Repair and Anti-Inflammatory Treatment of AKI-to-CKD Progression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310731. [PMID: 38805174 DOI: 10.1002/adma.202310731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 05/21/2024] [Indexed: 05/29/2024]
Abstract
As a central metabolic molecule, nicotinamide adenine dinucleotide (NAD+) can potentially treat acute kidney injury (AKI) and chronic kidney disease (CKD); however, its bioavailability is poor due to short half-life, instability, the deficiency of targeting, and difficulties in transmembrane transport. Here a physiologically adaptive gallic acid-NAD+ nanoparticle is designed, which has ultrasmall size and pH-responsiveness, passes through the glomerular filtration membrane to reach injured renal tubules, and efficiently delivers NAD+ into the kidneys. With an effective accumulation in the kidneys, it restores renal function, immune microenvironment homeostasis, and mitochondrial homeostasis of AKI mice via the NAD+-Sirtuin-1 axis, and exerts strong antifibrotic effects on the AKI-to-CKD transition by inhibiting TGF-β signaling. It also exhibits excellent stability, biodegradable, and biocompatible properties, ensuring its long-term safety, practicality, and clinical translational feasibility. The present study shows a potential modality of mitochondrial repair and immunomodulation through nanoagents for the efficient and safe treatment of AKI and CKD.
Collapse
Affiliation(s)
- Ying Kong
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, Shandong, China
| | - Xu Chen
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Feng Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, Shandong, China
| | - Jiageng Tang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Yijing Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiangxiang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Luyao Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Tong Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Yaqi Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Mengxiao Su
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Qixin Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Hanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Di Zhou
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Science, Shandong University, Jinan, 250012, Shandong, China
- National Key Laboratoy for innovation and Transfomation of Luobing Theoy, Key Laboratory of Cardiovascular Health, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, Shandong, China
- Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, Shandong, China
| | - Yi Fu
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| |
Collapse
|
12
|
Zhong J, Doughty R, Thymann T, Sangild PT, Nguyen DN, Muk T. Insulin-like growth factor-1 effects on kidney development in preterm piglets. Pediatr Res 2024:10.1038/s41390-024-03222-3. [PMID: 38762663 DOI: 10.1038/s41390-024-03222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Preterm birth disrupts fetal kidney development, potentially leading to postnatal acute kidney injury. Preterm infants are deficient in insulin-like growth factor 1 (IGF-1), a growth factor that stimulates organ development. By utilizing a preterm pig model, this study investigated whether IGF-1 supplementation enhances preterm kidney maturation. METHODS Cesarean-delivered preterm pigs were treated systemically IGF-1 or vehicle control for 5, 9 or 19 days after birth. Blood, urine, and kidney tissue were collected for biochemical, histological and gene expression analyses. Age-matched term-born pigs were sacrificed at similar postnatal ages and served as the reference group. RESULTS Compared with term pigs, preterm pigs exhibited impaired kidney maturation, as indicated by analyses of renal morphology, histopathology, and inflammatory and injury markers. Supplementation with IGF-1 reduced signs of kidney immaturity, particularly in the first week of life, as indicated by improved morphology, upregulated expression of key developmental genes, reduced severity and incidence of microscopic lesions, and decreased levels of inflammatory and injury markers. No association was seen between the symptoms of necrotizing enterocolitis and kidney defects. CONCLUSION Preterm birth in pigs impairs kidney maturation and exogenous IGF-1 treatment partially reverses this impairment. Early IGF-1 supplementation could support the development of preterm kidneys. IMPACT Preterm birth may disrupt kidney development in newborns, potentially leading to morphological changes, injury, and inflammation. Preterm pigs have previously been used as models for preterm infants, but not for kidney development. IGF-1 supplementation promotes kidney maturation and alleviates renal impairments in the first week of life in preterm pigs. IGF-1 may hold potential as a supportive therapy for preterm infants sensitive to acute kidney injury.
Collapse
Affiliation(s)
- Jingren Zhong
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Richard Doughty
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Thomas Thymann
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Per Torp Sangild
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Pediatrics, Odense University Hospital, Odense, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Tik Muk
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
13
|
Creed HA, Kannan S, Tate BL, Godefroy D, Banerjee P, Mitchell BM, Brakenhielm E, Chakraborty S, Rutkowski JM. Single-Cell RNA Sequencing Identifies Response of Renal Lymphatic Endothelial Cells to Acute Kidney Injury. J Am Soc Nephrol 2024; 35:549-565. [PMID: 38506705 PMCID: PMC11149045 DOI: 10.1681/asn.0000000000000325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/30/2024] [Indexed: 03/21/2024] Open
Abstract
SIGNIFICANCE STATEMENT The renal lymphatic vasculature and the lymphatic endothelial cells that make up this network play important immunomodulatory roles during inflammation. How lymphatics respond to AKI may affect AKI outcomes. The authors used single-cell RNA sequencing to characterize mouse renal lymphatic endothelial cells in quiescent and cisplatin-injured kidneys. Lymphatic endothelial cell gene expression changes were confirmed in ischemia-reperfusion injury and in cultured lymphatic endothelial cells, validating renal lymphatic endothelial cells single-cell RNA sequencing data. This study is the first to describe renal lymphatic endothelial cell heterogeneity and uncovers molecular pathways demonstrating lymphatic endothelial cells regulate the local immune response to AKI. These findings provide insights into previously unidentified molecular pathways for lymphatic endothelial cells and roles that may serve as potential therapeutic targets in limiting the progression of AKI. BACKGROUND The inflammatory response to AKI likely dictates future kidney health. Lymphatic vessels are responsible for maintaining tissue homeostasis through transport and immunomodulatory roles. Owing to the relative sparsity of lymphatic endothelial cells in the kidney, past sequencing efforts have not characterized these cells and their response to AKI. METHODS Here, we characterized murine renal lymphatic endothelial cell subpopulations by single-cell RNA sequencing and investigated their changes in cisplatin AKI 72 hours postinjury. Data were processed using the Seurat package. We validated our findings by quantitative PCR in lymphatic endothelial cells isolated from both cisplatin-injured and ischemia-reperfusion injury, by immunofluorescence, and confirmation in in vitro human lymphatic endothelial cells. RESULTS We have identified renal lymphatic endothelial cells and their lymphatic vascular roles that have yet to be characterized in previous studies. We report unique gene changes mapped across control and cisplatin-injured conditions. After AKI, renal lymphatic endothelial cells alter genes involved in endothelial cell apoptosis and vasculogenic processes as well as immunoregulatory signaling and metabolism. Differences between injury models were also identified with renal lymphatic endothelial cells further demonstrating changed gene expression between cisplatin and ischemia-reperfusion injury models, indicating the renal lymphatic endothelial cell response is both specific to where they lie in the lymphatic vasculature and the kidney injury type. CONCLUSIONS In this study, we uncover lymphatic vessel structural features of captured populations and injury-induced genetic changes. We further determine that lymphatic endothelial cell gene expression is altered between injury models. How lymphatic endothelial cells respond to AKI may therefore be key in regulating future kidney disease progression.
Collapse
Affiliation(s)
- Heidi A. Creed
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| | - Saranya Kannan
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| | - Brittany L. Tate
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| | - David Godefroy
- Inserm UMR1239 (Nordic Laboratory), UniRouen, Normandy University, Mont Saint Aignan, France
| | - Priyanka Banerjee
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| | - Brett M. Mitchell
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| | - Ebba Brakenhielm
- INSERM EnVI, UMR1096, University of Rouen Normandy, Rouen, France
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| | - Joseph M. Rutkowski
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| |
Collapse
|
14
|
Smith T, Zaidi A, Brown CVM, Pino-Chavez G, Bowen T, Meran S, Fraser D, Chavez R, Khalid U. Robust Rat and Mouse Models of Bilateral Renal Ischemia Reperfusion Injury. In Vivo 2024; 38:1049-1057. [PMID: 38688639 PMCID: PMC11059907 DOI: 10.21873/invivo.13538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND/AIM Acute and chronic kidney diseases are a major contributor to morbidity and mortality worldwide, with no specific treatments currently available for these. To enable understanding the pathophysiology of and testing novel treatments for acute and chronic kidney disease, a suitable in vivo model of kidney disease is essential. In this article, we describe two reliable rodent models (rats and mice) of efficacious kidney injury displaying acute to chronic kidney injury progression, which is also reversible through novel therapeutic strategies such as ischemic preconditioning (IPC). MATERIALS AND METHODS We utilized adult male Lewis rats and adult male wildtype (C57BL/6) mice, performed a midline laparotomy, and induced warm ischemia to both kidneys by bilateral clamping of both renal vascular pedicles for a set time, to mimic the hypoxic etiology of disease commonly found in kidney injury. RESULTS Bilateral ischemia reperfusion injury caused marked structural and functional kidney injury as exemplified by histology damage scores, serum creatinine levels, and kidney injury biomarker levels in both rodents. Furthermore, this effect displayed a dose-dependent response in the mouse model. CONCLUSION These rodent models of bilateral kidney IRI are reliable, reproducible, and enable detailed mechanistic study of the underlying pathophysiology of both acute and chronic kidney disease. They have been carefully optimised for single operator use with a strong track record of training both surgically trained and surgically naïve operators.
Collapse
Affiliation(s)
- Tanya Smith
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K.;
- Department of Anesthetics, Cardiff & Vale University Health Board, University Hospital of Wales, Cardiff, U.K
| | - Aeliya Zaidi
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
- Cardiff Transplant Unit, Nephrology & Transplant Directorate, Cardiff & Vale University Health Board, University Hospital of Wales, Cardiff, U.K
| | - Charlotte Victoria Maynard Brown
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
- Cardiff Transplant Unit, Nephrology & Transplant Directorate, Cardiff & Vale University Health Board, University Hospital of Wales, Cardiff, U.K
| | - Gilda Pino-Chavez
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| | - Timothy Bowen
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| | - Soma Meran
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| | - Donald Fraser
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| | - Rafael Chavez
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
- Cardiff Transplant Unit, Nephrology & Transplant Directorate, Cardiff & Vale University Health Board, University Hospital of Wales, Cardiff, U.K
| | - Usman Khalid
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K.;
- Cardiff Transplant Unit, Nephrology & Transplant Directorate, Cardiff & Vale University Health Board, University Hospital of Wales, Cardiff, U.K
| |
Collapse
|
15
|
Inoue T, Umene R, Sung SSJ, Tanaka S, Huang L, Yao J, Hashimoto N, Wu CH, Nakamura Y, Nishino T, Ye H, Rosin DL, Ishihara K, Okusa MD. Bone marrow stromal cell antigen-1 deficiency protects from acute kidney injury. Am J Physiol Renal Physiol 2024; 326:F167-F177. [PMID: 37969103 DOI: 10.1152/ajprenal.00175.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/16/2023] [Accepted: 11/01/2023] [Indexed: 11/17/2023] Open
Abstract
This study aimed to investigate the role of bone marrow stromal cell antigen-1 (Bst1; also known as CD157) in acute kidney injury (AKI). Bst1 is a cell surface molecule with various enzymatic activities and downstream intracellular signaling pathways that modulate the immune response. Previous research has linked Bst1 to diseases such as ovarian cancer, Parkinson's disease, and rheumatoid arthritis. We used bilateral ischemia-reperfusion injury (IRI) as an AKI model and created bone marrow chimeric mice to evaluate the role of Bst1 in bone marrow-derived cells. We also used flow cytometry to identify Bst1/CD157 expression in hematopoietic cells and evaluate immune cell dynamics in the kidney. The findings showed that Bst1-deficient (Bst1-/-) mice were protected against renal bilateral IRI. Bone marrow chimera experiments revealed that Bst1 expression on hematopoietic cells, but not parenchymal cells, induced renal IRI. Bst1 was mainly found in B cells and neutrophils by flow cytometry of the spleen and bone marrow. In vitro, migration of neutrophils from Bst1-/- mice was suppressed, and adoptive transfer of neutrophils from wild-type Bst1+/+ mice abolished the renal protective effect in Bst1 knockout mice. In conclusion, the study demonstrated that Bst1-/- mice are protected against renal IRI and that Bst1 expression in neutrophils plays a crucial role in inducing renal IRI. These findings suggest that targeting Bst1 in neutrophils could be a potential therapeutic strategy for AKI.NEW & NOTEWORTHY Acute kidney injury (AKI), a serious disease for which there is no effective Federal Drug Administration-approved treatment, is associated with high mortality rates. Bone marrow stromal cell antigen-1 (Bst1) is a cell surface molecule that can cause kidney fibrosis, but its role in AKI is largely unknown. Our study showed that Bst1-/- mice revealed a protective effect against renal bilateral ischemia-reperfusion injury (IRI). Adoptive transfer studies confirmed that Bst1 expression in hematopoietic cells, especially neutrophils, contributed to renal bilateral IRI.
Collapse
Affiliation(s)
- Tsuyoshi Inoue
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, United States
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Ryusuke Umene
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Sun-Sang J Sung
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Shinji Tanaka
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Liping Huang
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Junlan Yao
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Noritatsu Hashimoto
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Chia-Hsien Wu
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yasuna Nakamura
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tomoya Nishino
- Department of Nephrology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hong Ye
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Diane L Rosin
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States
| | - Katsuhiko Ishihara
- Department of Design for Medical and Health Care, Faculty of Health and Welfare Services Administration, Kawasaki University of Medical Welfare, Okayama, Japan
| | - Mark D Okusa
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
16
|
Nagata C, Hata M, Miyazaki Y, Masuda H, Wada T, Kimura T, Fujii M, Sakurai Y, Matsubara Y, Yoshida K, Miyagawa S, Ikeda M, Ueno T. Development of postoperative delirium prediction models in patients undergoing cardiovascular surgery using machine learning algorithms. Sci Rep 2023; 13:21090. [PMID: 38036664 PMCID: PMC10689441 DOI: 10.1038/s41598-023-48418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023] Open
Abstract
Associations between delirium and postoperative adverse events in cardiovascular surgery have been reported and the preoperative identification of high-risk patients of delirium is needed to implement focused interventions. We aimed to develop and validate machine learning models to predict post-cardiovascular surgery delirium. Patients aged ≥ 40 years who underwent cardiovascular surgery at a single hospital were prospectively enrolled. Preoperative and intraoperative factors were assessed. Each patient was evaluated for postoperative delirium 7 days after surgery. We developed machine learning models using the Bernoulli naive Bayes, Support vector machine, Random forest, Extra-trees, and XGBoost algorithms. Stratified fivefold cross-validation was performed for each developed model. Of the 87 patients, 24 (27.6%) developed postoperative delirium. Age, use of psychotropic drugs, cognitive function (Mini-Cog < 4), index of activities of daily living (Barthel Index < 100), history of stroke or cerebral hemorrhage, and eGFR (estimated glomerular filtration rate) < 60 were selected to develop delirium prediction models. The Extra-trees model had the best area under the receiver operating characteristic curve (0.76 [standard deviation 0.11]; sensitivity: 0.63; specificity: 0.78). XGBoost showed the highest sensitivity (AUROC, 0.75 [0.07]; sensitivity: 0.67; specificity: 0.79). Machine learning algorithms could predict post-cardiovascular delirium using preoperative data.Trial registration: UMIN-CTR (ID; UMIN000049390).
Collapse
Affiliation(s)
- Chie Nagata
- Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Masahiro Hata
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuki Miyazaki
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirotada Masuda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tamiki Wada
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tasuku Kimura
- SANKEN (The Institution of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka, Japan
| | - Makoto Fujii
- Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasushi Sakurai
- SANKEN (The Institution of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka, Japan
| | - Yasuko Matsubara
- SANKEN (The Institution of Scientific and Industrial Research), Osaka University, Ibaraki, Osaka, Japan
| | - Kiyoshi Yoshida
- Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takayoshi Ueno
- Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
17
|
Yan YT, Liu HM, Kong YF, Liu JM, Li C, Zhao BC, Liu KX. Association of preoperative neutrophil-lymphocyte ratio with acute kidney injury in patients with non-cardiac surgery: difference among surgical types. Int Urol Nephrol 2023; 55:2647-2656. [PMID: 36964822 DOI: 10.1007/s11255-023-03567-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 03/18/2023] [Indexed: 03/26/2023]
Abstract
PURPOSE To examine the relationship between Neutrophil-Lymphocyte Ratio (NLR) and Acute Kidney Injury (AKI) in patients undergoing noncardiac surgery, and subgroup analysis was performed for different types of non-cardiac surgery. METHODS The present retrospective cohort study included 10,159 adult patients who underwent major noncardiac surgery at Nanfang Hospital, Southern Medical University, between 2008 and 2018. Postoperative AKI was defined as an increase in serum creatinine level of at least 0.3 mg/dl within 48 h, or 1.5 times higher than baseline within 7 days postoperatively according to the Kidney Disease Improving Global Outcome. The correlation between preoperative NLR and postoperative AKI was determined by stepwise multivariate logistic regression analysis, and the predictive value of NLR was evaluated by the receiver operating characteristics curve (ROC) analysis. RESULTS Four hundred and eighty-five (4.77%) patients developed AKI postoperatively. Preoperative NLR was independently associated with postoperative AKI in all patients undergoing non-cardiac surgery (Odds ratio [OR], 1.03; 95% confidence interval [CI], 1.00-1.06). The optimal cut-off value of NLR was 2.12 according ROC analysis. The OR and 95% CI of AKI for NLR > 2.12 was 1.48 (1.21-1.81) compared with NLR ≤ 2.12. In addition, the positive association was mainly shown in patients undergone digestive system surgery with a cut-off value of 2.12 but not in neurological and musculoskeletal system surgeries. CONCLUSION The present study confirmed the association of preoperative NLR with postoperative AKI in digestive system surgical patients. A NLR value of 2.12 may be a useful cut-off to evaluate the risk of AKI.
Collapse
Affiliation(s)
- Yang-Tian Yan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hua-Min Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi-Fan Kong
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia-Ming Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bing-Cheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Li L, Wang S, Wang W. Knockdown of ELF4 aggravates renal injury in ischemia/reperfusion mice through promotion of pyroptosis, inflammation, oxidative stress, and endoplasmic reticulum stress. BMC Mol Cell Biol 2023; 24:22. [PMID: 37474923 PMCID: PMC10360327 DOI: 10.1186/s12860-023-00485-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Renal ischemia/reperfusion (I/R) injury is a major cause of acute kidney injury (AKI). Dysfunction of E74-like ETS transcription factor 4 (ELF4) leads to inflammation. This research intended to look into the function and mechanisms of ELF4 in I/R and oxygen-glucose deprivation/reperfusion (OGD/R) model. RESULTS In I/R and OGD/R model, ELF4 expression was downregulated. ELF4 knockout aggravated I/R-induced kidney injury, oxidative stress (OS), endoplasmic reticulum stress (ERS), apoptosis, inflammation, and pyroptosis in mice. In HK-2 cells treated with OGD/R, suppression of ELF4 expression inhibited cell proliferation and promoted cell apoptosis, OS, ERS, inflammation, and pyroptosis. Moreover, ELF4 overexpression led to the opposite results. CONCLUSION ELF4 deficiency aggravated I/R induced AKI, which was involved in apoptosis, OS, ERS, inflammation, and pyroptosis. Targeting ELF4 may be a promising new therapeutic strategy for preventing inflammation after IR-AKI.
Collapse
Affiliation(s)
- Li Li
- Department of Nephrology, Jinan City People's Hospital, No. 001, Changshao North Road, Laiwu District, Jinan, Shandong, 271199, People's Republic of China.
| | - Shunying Wang
- Department of Cadre Health Section, Jinan City People's Hospital, Jinan, Shandong, 271199, People's Republic of China
| | - Wenming Wang
- Department of Cadre Health Section, Jinan City People's Hospital, Jinan, Shandong, 271199, People's Republic of China
| |
Collapse
|
19
|
Creed HA, Kannan S, Tate BL, Banerjee P, Mitchell BM, Chakraborty S, Rutkowski JM. Single-cell RNA sequencing identifies response of renal lymphatic endothelial cells to acute kidney injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.09.544380. [PMID: 37333313 PMCID: PMC10274866 DOI: 10.1101/2023.06.09.544380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The inflammatory response to acute kidney injury (AKI) likely dictates future renal health. Lymphatic vessels are responsible for maintaining tissue homeostasis through transport and immunomodulatory roles. Due to the relative sparsity of lymphatic endothelial cells (LECs) in the kidney, past sequencing efforts have not characterized these cells and their response to AKI. Here we characterized murine renal LEC subpopulations by single-cell RNA sequencing and investigated their changes in cisplatin AKI. We validated our findings by qPCR in LECs isolated from both cisplatin-injured and ischemia reperfusion injury, by immunofluorescence, and confirmation in in vitro human LECs. We have identified renal LECs and their lymphatic vascular roles that have yet to be characterized in previous studies. We report unique gene changes mapped across control and cisplatin injured conditions. Following AKI, renal LECs alter genes involved endothelial cell apoptosis and vasculogenic processes as well as immunoregulatory signaling and metabolism. Differences between injury models are also identified with renal LECs further demonstrating changed gene expression between cisplatin and ischemia reperfusion injury models, indicating the renal LEC response is both specific to where they lie in the lymphatic vasculature and the renal injury type. How LECs respond to AKI may therefore be key in regulating future kidney disease progression.
Collapse
|
20
|
Wang Q, Li S, Sun M, Ma J, Sun J, Fan M. Systemic immune-inflammation index may predict the acute kidney injury and prognosis in patients with spontaneous cerebral hemorrhage undergoing craniotomy: a single-center retrospective study. BMC Nephrol 2023; 24:73. [PMID: 36964487 PMCID: PMC10039500 DOI: 10.1186/s12882-023-03124-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/20/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND The systemic immune-inflammation index (SII) is an emerging prognostic marker of cancer. We aimed to explore the predictive ability of the SII on acute kidney injury (AKI) and prognosis in patients with spontaneous cerebral hemorrhage (SCH) who underwent craniotomy. METHODS Patients with SCH who underwent craniotomy between 2014 and 2021 were enrolled in this study. The epidemiology and predictive factors for AKI after SCH were analyzed. The prognostic factors for clinical outcomes in patients with SCH and AKI were further investigated. The prognostic factors were then analyzed using a logistic regression model and a receiver operating characteristic curve. RESULTS In total, 305 patients were enrolled in this study. Of these, 129 (42.3%) patients presented with AKI, and 176 (57.7%) patients were unremarkable. The SII (odds ratio [OR], 1.261; 95% confidence interval [CI], 1.036-1.553; P = 0.020) values and serum uric acid levels (OR, 1.004; 95% CI, 1.001-1.007; P = 0.005) were significant predictors of AKI after SCH craniotomy. The SII cutoff value was 1794.43 (area under the curve [AUC], 0.669; 95% CI, 0.608-0.730; P < 0.001; sensitivity, 65.9%; specificity, 65.1%). Of the patients with AKI, 95 and 34 achieved poor and good outcomes, respectively. SII values (OR, 2.667; 95% CI, 1.167-6.095; P = 0.020), systemic inflammation response index values (OR, 1.529; 95% CI, 1.064-2.198; P = 0.022), and Glasgow Coma Scale (GCS) scores on admission (OR, 0.593; 95% CI, 0.437-0.805; P = 0.001) were significant in the multivariate logistic regression analysis. The cutoff SII value was 2053.51 (AUC, 0.886; 95% CI, 0.827-0.946; P < 0.001; sensitivity, 78.9%; specificity, 88.2%). CONCLUSIONS The SII may predict AKI in patients with SCH who underwent craniotomy and may also predict the short-term prognosis of these patients.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Nephrology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Shifang Li
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Meifeng Sun
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junwei Ma
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jian Sun
- Department of Neurosurgical Intensive Care Unit, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingchao Fan
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- Department of Neurosurgical Intensive Care Unit, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
21
|
Conroy AL, Datta D, Hoffmann A, Wassmer SC. The kidney-brain pathogenic axis in severe falciparum malaria. Trends Parasitol 2023; 39:191-199. [PMID: 36737313 PMCID: PMC11071448 DOI: 10.1016/j.pt.2023.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/05/2023]
Abstract
Severe falciparum malaria is a medical emergency and a leading cause of death and neurodisability in endemic areas. Common complications include acute kidney injury (AKI) and cerebral malaria, and recent studies have suggested links between kidney and brain dysfunction in Plasmodium falciparum infection. Here, we review these new findings and present the hypothesis of a pivotal pathogenic crosstalk between the kidneys and the brain in severe falciparum malaria. We highlight the evidence of a role for distant organ involvement in the development of cerebral malaria and subsequent neurocognitive impairment post-recovery, describe the challenges associated with current diagnostic shortcomings for both AKI and brain involvement in severe falciparum malaria, and explore novel potential therapeutic strategies.
Collapse
Affiliation(s)
- Andrea L Conroy
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Dibyadyuti Datta
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Angelika Hoffmann
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Samuel C Wassmer
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
22
|
Brezgunova AA, Andrianova NV, Popkov VA, Tkachev SY, Manskikh VN, Pevzner IB, Zorova LD, Timashev PS, Silachev DN, Zorov DB, Plotnikov EY. New experimental model of kidney injury: Photothrombosis-induced kidney ischemia. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166622. [PMID: 36526237 DOI: 10.1016/j.bbadis.2022.166622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) is a frequent pathology with a high mortality rate after even a single AKI episode and a great risk of chronic kidney disease (CKD) development. To get insight into mechanisms of the AKI pathogenesis, there is a need to develop diverse experimental models of the disease. Photothrombosis is a widely used method for inducing ischemia in the brain. In this study, for the first time, we described photothrombosis-induced kidney ischemia as an appropriate model of AKI and obtained comprehensive characteristics of the photothrombotic lesion using micro-computed tomography (micro-CT) and histological techniques. In the ischemic area, we observed destruction of tubules, the loss of brush border and nuclei, connective tissue fibers disorganization, leukocyte infiltration, and hyaline casts formation. In kidney tissue and urine, we revealed increased levels in markers of proliferation and injury. The explicit long-term consequence of photothrombosis-induced kidney ischemia was renal fibrosis. Thus, we establish a new low invasive experimental model of AKI, which provides a reproducible local ischemic injury lesion. We propose our model of photothrombosis-induced kidney ischemia as a useful approach for investigating AKI pathogenesis, studying the mechanisms of kidney regeneration, and development of therapy against AKI and CKD.
Collapse
Affiliation(s)
- Anna A Brezgunova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia; A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Nadezda V Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vasily A Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Sergey Y Tkachev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Vasily N Manskikh
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Irina B Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Ljubava D Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Peter S Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Denis N Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Dmitry B Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia.
| | - Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia.
| |
Collapse
|
23
|
Oral hydrogel microspheres were used for highly specific delivery of Steamed Codonopsis lanceolata to exert the protective effects on cisplatin-induced acute kidney injury in mice. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
24
|
Yan P, Duan SB, Luo XQ, Zhang NY, Deng YH. Development and validation of a deep neural network-based model to predict acute kidney injury following intravenous administration of iodinated contrast media in hospitalized patients with chronic kidney disease: a multicohort analysis. Nephrol Dial Transplant 2023; 38:352-361. [PMID: 35218197 DOI: 10.1093/ndt/gfac049] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Stratification of chronic kidney disease (CKD) patients [estimated glomerular filtration rate (eGFR) <60 mL/min/1.73 m2] at risk for post-contrast acute kidney injury (PC-AKI) following intravenous administration of iodinated contrast media (ICM) is important for clinical decision-making and clinical trial enrollment. METHODS The derivation and internal validation cohorts originated from the Second Xiangya Hospital. The external validation cohort was generated from the Xiangya Hospital and the openly accessible database Medical Information Mart for Intensive CareIV. PC-AKI was defined based on the serum creatinine criteria of the Kidney Disease: Improving Global Outcomes (KDIGO). Six feature selection methods were used to identify the most influential predictors from 79 candidate variables. Deep neural networks (DNNs) were used to establish the model and compared with logistic regression analyses. Model discrimination was evaluated by area under the receiver operating characteristic curve (AUC). Low-risk and high-risk cutoff points were set to stratify patients. RESULTS Among 4218 encounters studied, PC-AKI occurred in 10.3, 10.4 and 11.4% of encounters in the derivation, internal and external validation cohorts, respectively. The 14 variables-based DNN model had significantly better performance than the logistic regression model with AUC being 0.939 (95% confidence interval: 0.916-0.958) and 0.940 (95% confidence interval: 0.909-0.954) in the internal and external validation cohorts, respectively, and showed promising discrimination in subgroup analyses (AUC ≥ 0.800). The observed PC-AKI risks increased significantly from the low- to intermediate- to high-risk group (<1.0 to >50%) and the accuracy of patients not developing PC-AKI was 99% in the low-risk category in both the internal and external validation cohorts. CONCLUSIONS A DNN model using routinely available variables can accurately discriminate the risk of PC-AKI of hospitalized CKD patients following intravenous administration of ICM.
Collapse
Affiliation(s)
- Ping Yan
- Department of Nephrology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Shao-Bin Duan
- Department of Nephrology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Xiao-Qin Luo
- Department of Nephrology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Ning-Ya Zhang
- Information Center, The Second Xiangya Hospital of Central South University; Changsha, Hunan, China
| | - Ying-Hao Deng
- Department of Nephrology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
25
|
Vernier ICS, Neres-Santos RS, Andrade-Oliveira V, Carneiro-Ramos MS. Immune Cells Are Differentially Modulated in the Heart and the Kidney during the Development of Cardiorenal Syndrome 3. Cells 2023; 12:605. [PMID: 36831272 PMCID: PMC9953884 DOI: 10.3390/cells12040605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/28/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Cardiorenal syndrome type 3 (CRS 3) occurs when there is an acute kidney injury (AKI) leading to the development of an acute cardiac injury. The immune system is involved in modulating the severity of kidney injury, and the role of immune system cells in the development of CRS 3 is not well established. The present work aims to characterize the macrophage and T and B lymphocyte populations in kidney and heart tissue after AKI induced by renal I/R. Thus, C57BL/6 mice were subjected to a renal I/R protocol by occlusion of the left renal pedicle (unilateral) for 60 min, followed by reperfusion for 3, 8 and 15 days. The immune cell populations of interest were identified using flow cytometry, and RT-qPCR was used to evaluate gene expression. As a result, a significant increase in TCD4+, TCD8+ lymphocytes and M1 macrophages to the renal tissue was observed, while B cells in the heart decreased. A renal tissue repair response characterized by Foxp3 activation predominated. However, a more inflammatory profile was shown in the heart tissue influenced by IL-17RA and IL-1β. In conclusion, the AKI generated by renal I/R was able to activate and recruit T and B lymphocytes and macrophages, as well as pro-inflammatory mediators to renal and cardiac tissue, showing the role of the immune system as a bridge between both organs in the context of CRS 3.
Collapse
Affiliation(s)
- Imara Caridad Stable Vernier
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, São Paulo 09210-580, Brazil
| | - Raquel Silva Neres-Santos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, São Paulo 09210-580, Brazil
| | - Vinicius Andrade-Oliveira
- Bernardo’s Laboratory, Center of Natural and Human Sciences (CCNH), Federal University of ABC, São Paulo 09210-580, Brazil
| | - Marcela Sorelli Carneiro-Ramos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, São Paulo 09210-580, Brazil
| |
Collapse
|
26
|
Vallés PG, Gil Lorenzo AF, Garcia RD, Cacciamani V, Benardon ME, Costantino VV. Toll-like Receptor 4 in Acute Kidney Injury. Int J Mol Sci 2023; 24:ijms24021415. [PMID: 36674930 PMCID: PMC9864062 DOI: 10.3390/ijms24021415] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023] Open
Abstract
Acute kidney injury (AKI) is a common and devastating pathologic condition, associated with considerable high morbidity and mortality. Although significant breakthroughs have been made in recent years, to this day no effective pharmacological therapies for its treatment exist. AKI is known to be connected with intrarenal and systemic inflammation. The innate immune system plays an important role as the first defense response mechanism to tissue injury. Toll-like receptor 4 (TLR4) is a well-characterized pattern recognition receptor, and increasing evidence has shown that TLR4 mediated inflammatory response, plays a pivotal role in the pathogenesis of acute kidney injury. Pathogen-associated molecular patterns (PAMPS), which are the conserved microbial motifs, are sensed by these receptors. Endogenous molecules generated during tissue injury, and labeled as damage-associated molecular pattern molecules (DAMPs), also activate pattern recognition receptors, thereby offering an understanding of sterile types of inflammation. Excessive, uncontrolled and/or sustained activation of TLR4, may lead to a chronic inflammatory state. In this review we describe the role of TLR4, its endogenous ligands and activation in the inflammatory response to ischemic/reperfusion-induced AKI and sepsis-associated AKI. The potential regeneration signaling patterns of TLR4 in acute kidney injury, are also discussed.
Collapse
Affiliation(s)
- Patricia G. Vallés
- Área de Fisiopatología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, Mendoza 5500, Argentina
- IMBECU-CONICET (Instituto de Medicina y Biología Experimental de Cuyo—Consejo Nacional de Investigaciones Científicas y Técnicas), Mendoza 5500, Argentina
- Correspondence:
| | - Andrea Fernanda Gil Lorenzo
- Área de Fisiopatología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, Mendoza 5500, Argentina
| | - Rodrigo D. Garcia
- Área de Fisiopatología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, Mendoza 5500, Argentina
| | - Valeria Cacciamani
- IMBECU-CONICET (Instituto de Medicina y Biología Experimental de Cuyo—Consejo Nacional de Investigaciones Científicas y Técnicas), Mendoza 5500, Argentina
| | - María Eugenia Benardon
- Área de Fisiopatología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, Mendoza 5500, Argentina
| | - Valeria Victoria Costantino
- IMBECU-CONICET (Instituto de Medicina y Biología Experimental de Cuyo—Consejo Nacional de Investigaciones Científicas y Técnicas), Mendoza 5500, Argentina
- Área de Biología Celular, Departamento de Morfofisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, Mendoza 5500, Argentina
| |
Collapse
|
27
|
Silva AVBDA, Campanati JDEAG, Barcelos IDES, Santos ACL, Deus UPDE, Soares TDEJ, Amaral LSDEB. COVID-19 and Acute Kidney Injury - Direct and Indirect Pathophysiological Mechanisms Underlying Lesion Development. AN ACAD BRAS CIENC 2022; 94:e20211501. [PMID: 36477239 DOI: 10.1590/0001-3765202220211501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/08/2022] [Indexed: 11/29/2022] Open
Abstract
COVID-19 is a pandemic disease caused by the SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) responsible for millions of deaths worldwide. Although the respiratory system is the main target of COVID-19, the disease can affect other organs, including the kidneys. Acute Kidney Injury (AKI), commonly seen in patients infected with COVID-19, has a multifactorial cause. Several studies associate this injury with the direct involvement of the virus in renal cells and the indirect damage stimulated by the infection. The direct cytopathic effects of SARS-CoV-2 are due to the entry and replication of the virus in renal cells, changing several regulatory pathways, especially the renin-angiotensin-aldosterone system (RAAS), with repercussions on the kallikrein-kinin system (KKS). Furthermore, the virus can deregulate the immune system, leading to an exaggerated response of inflammatory cells, characterizing the state of hypercytokinemia. The such exaggerated inflammatory response is commonly associated with hemodynamic changes, reduced renal perfusion, tissue hypoxia, generation of reactive oxygen species (ROS), endothelial damage, and coagulopathies, which can result in severe damage to the renal parenchyma. Thereby, understanding the molecular mechanisms and pathophysiology of kidney injuries induced by SARS-COV-2 is of fundamental importance to obtaining new therapeutic insights for the prevention and management of AKI.
Collapse
Affiliation(s)
- Antônio V B DA Silva
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - João DE A G Campanati
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Isadora DE S Barcelos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Alberto C L Santos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Uildson P DE Deus
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Telma DE J Soares
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| | - Liliany S DE B Amaral
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, 45029-094 Vitória da Conquista, BA, Brazil
| |
Collapse
|
28
|
Nie Y, Wang L, You X, Wang X, Wu J, Zheng Z. Low dimensional nanomaterials for treating acute kidney injury. J Nanobiotechnology 2022; 20:505. [PMID: 36456976 PMCID: PMC9714216 DOI: 10.1186/s12951-022-01712-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022] Open
Abstract
Acute kidney injury (AKI) is one of the most common severe complications among hospitalized patients. In the absence of specific drugs to treat AKI, hemodialysis remains the primary clinical treatment for AKI patients. AKI treatment has received significant attention recently due to the excellent drug delivery capabilities of low-dimensional nanomaterials (LDNs) and their unique therapeutic effects. Diverse LDNs have been proposed to treat AKI, with promising results and the potential for future clinical application. This article aims to provide an overview of the pathogenesis of AKI and the recent advances in the treatment of AKI using different types of LDNs. In addition, it is intended to provide theoretical support for the design of LDNs and implications for AKI treatment.
Collapse
Affiliation(s)
- Yuanpeng Nie
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Liying Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinru You
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaohua Wang
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, China
| | - Jun Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, China.
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
29
|
Novel gold-platinum nanoparticles serve as broad-spectrum antioxidants for attenuating ischemia reperfusion injury of the kidney. Kidney Int 2022; 102:1057-1072. [PMID: 35870640 DOI: 10.1016/j.kint.2022.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/22/2022] [Accepted: 07/06/2022] [Indexed: 02/05/2023]
Abstract
Kidney ischemia reperfusion injury (IRI) is a common and inevitable pathological condition in routine urological practices, especially during transplantation. Severe kidney IRI may even induce systemic damage to peripheral organs, and lead to multisystem organ failure. However, no standard clinical treatment option is currently available. It has been reported that kidney IRI is predominantly associated with abnormally increased endogenous reactive oxygen species (ROS). Scavenging excessive ROS may reduce the damage caused by oxidative stress and subsequently alleviate kidney IRI. Here, we reported a simple and efficient one-step synthesis of gold-platinum nanoparticles (AuPt NPs) with a gold core having a loose and branched outer platinum shell with superior ROS scavenging capacity to possibly treat kidney IRI. These AuPt NPs exhibited multiple enzyme-like anti-oxidative properties simultaneously possessing catalase- and peroxidase-like activity. These particles showed excellent cell protective capability, and alleviated kidney IRI both in vitro and in vivo without obvious toxicity, by suppressing cell apoptosis, inflammatory cytokine release, and inflammasome formation. Meanwhile, AuPt NPs also had an effect on inhibiting the transition to chronic kidney disease by reducing kidney fibrosis in the long term. Thus, AuPt NPs might be a good therapeutic agent for kidney IRI management and may be helpful for the development of clinical treatments for kidney IRI.
Collapse
|
30
|
Borges FK, Devereaux PJ, Cuerden M, Sontrop JM, Bhandari M, Guerra-Farfán E, Patel A, Sigamani A, Umer M, Neary J, Tiboni M, Tandon V, Ramokgopa MT, Sancheti P, Lawendy AR, Balaguer-Castro M, Jenkinson R, Ślęczka P, Nur AN, Wood GCA, Feibel RJ, McMahon JS, Biccard BM, Ortalda A, Szczeklik W, Wang CY, Tomás-Hernández J, Vincent J, Harvey V, Pettit S, Balasubramanian K, Slobogean G, Garg AX. Accelerated Surgery Versus Standard Care in Hip Fracture (HIP ATTACK-1): A Kidney Substudy of a Randomized Clinical Trial. Am J Kidney Dis 2022; 80:686-689. [PMID: 35346742 DOI: 10.1053/j.ajkd.2022.01.431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/14/2022] [Indexed: 02/02/2023]
Affiliation(s)
- Flavia K Borges
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Perioperative Medicine, Population Health Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - P J Devereaux
- Department of Perioperative Medicine, Population Health Research Institute, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Meaghan Cuerden
- Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada
| | - Jessica M Sontrop
- Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada; Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | - Mohit Bhandari
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Ernesto Guerra-Farfán
- Department of Orthopaedic Surgery and Traumatology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Ameen Patel
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Perioperative Medicine, Population Health Research Institute, Hamilton, Ontario, Canada
| | - Alben Sigamani
- Clinical Research, Narayana Hrudayalaya Limited, Bangalore, India
| | - Masood Umer
- Department of Orthopaedic Surgery, Aga Khan University, Karachi, Pakistan
| | - John Neary
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Maria Tiboni
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Vikas Tandon
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mmampapatla Thomas Ramokgopa
- Department of Orthopaedic Surgery, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa; Department of Orthopaedic Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - Parag Sancheti
- Department of Orthopaedic Surgery, Sancheti Institute for Orthopaedics & Rehabilitation, Pune, India
| | | | - Mariano Balaguer-Castro
- Department of Orthopaedic Surgery and Traumatology, Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Richard Jenkinson
- Department of Surgery and Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | | - Aamer Nabi Nur
- Department of Orthopaedic Surgery, Shifa International Hospital, Islamabad, Pakistan
| | - Gavin C A Wood
- Department of Surgery, Queen's University, Kingston, Ontario, Canada
| | - Robert J Feibel
- Division of Orthopaedic Surgery, Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Bruce M Biccard
- Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Observatory, South Africa
| | - Alessandro Ortalda
- Department of Anaesthesia and Intensive Care, Vita-Salute San Raffaele University, Milan, Italy
| | - Wojciech Szczeklik
- Center for Intensive Care and Perioperative Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Chew Yin Wang
- Department of Anaesthesiology, University of Malaya, Faculty of Medicine, Kuala Lumpur, Malaysia
| | - Jordi Tomás-Hernández
- Department of Orthopaedic Surgery and Traumatology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Jessica Vincent
- Department of Perioperative Medicine, Population Health Research Institute, Hamilton, Ontario, Canada
| | - Valerie Harvey
- Department of Perioperative Medicine, Population Health Research Institute, Hamilton, Ontario, Canada
| | - Shirley Pettit
- Department of Perioperative Medicine, Population Health Research Institute, Hamilton, Ontario, Canada
| | - Kumar Balasubramanian
- Department of Perioperative Medicine, Population Health Research Institute, Hamilton, Ontario, Canada
| | - Gerard Slobogean
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Amit X Garg
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada; Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada; Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada.
| | | |
Collapse
|
31
|
Zhu Y, Yang S, Lv L, Zhai X, Wu G, Qi X, Dong D, Tao X. Research Progress on the Positive and Negative Regulatory Effects of Rhein on the Kidney: A Review of Its Molecular Targets. Molecules 2022; 27:molecules27196572. [PMID: 36235108 PMCID: PMC9573519 DOI: 10.3390/molecules27196572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Currently, both acute kidney injury (AKI) and chronic kidney disease (CKD) are considered to be the leading public health problems with gradually increasing incidence rates around the world. Rhein is a monomeric component of anthraquinone isolated from rhubarb, a traditional Chinese medicine. It has anti-inflammation, anti-oxidation, anti-apoptosis, anti-bacterial and other pharmacological activities, as well as a renal protective effects. Rhein exerts its nephroprotective effects mainly through decreasing hypoglycemic and hypolipidemic, playing anti-inflammatory, antioxidant and anti-fibrotic effects and regulating drug-transporters. However, the latest studies show that rhein also has potential kidney toxicity in case of large dosages and long use times. The present review highlights rhein's molecular targets and its different effects on the kidney based on the available literature and clarifies that rhein regulates the function of the kidney in a positive and negative way. It will be helpful to conduct further studies on how to make full use of rhein in the kidney and to avoid kidney damage so as to make it an effective kidney protection drug.
Collapse
|
32
|
Juncos LA, Wieruszewski PM, Kashani K. Pathophysiology of Acute Kidney Injury in Critical Illness: A Narrative Review. Compr Physiol 2022; 12:3767-3780. [PMID: 36073750 DOI: 10.1002/cphy.c210028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute kidney injury (AKI) is a syndrome that entails a rapid decline in kidney function with or without injury. The consequences of AKI among acutely ill patients are dire and lead to higher mortality, morbidity, and healthcare cost. To prevent AKI and its short and long-term repercussions, understanding its pathophysiology is essential. Depending on the baseline kidney histology and function reserves, the number of kidney insults, and the intensity of each insult, the clinical presentation of AKI may differ. While many factors are capable of inducing renal injury, they can be categorized into a few processes. The three primary processes reported in the literature are hemodynamic changes, inflammatory reactions, and nephrotoxicity. The majority of patients with AKI will suffer from more than one during their development and/or progression of AKI. Moreover, the development of one usually leads to the instigation of another. Thus, the interactions and progression between these mechanisms may determine the severity and duration of the AKI. Other factors such as organ crosstalk and how our concurrent therapies interact with these mechanisms complicate the pathophysiology of the progression of the AKI even further. In this narrative review article, we describe these three main pathophysiological processes that lead to the development and progression of AKI. © 2022 American Physiological Society. Compr Physiol 12: 1-14, 2022.
Collapse
Affiliation(s)
- Luis A Juncos
- Division of Nephrology, Central Arkansas Veterans' Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Patrick M Wieruszewski
- Division of Hospital Pharmacy, Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Infection in Living Donor Liver Transplantation Leads to Increased Risk of Adverse Renal Outcomes. Nutrients 2022; 14:nu14173660. [PMID: 36079917 PMCID: PMC9460461 DOI: 10.3390/nu14173660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022] Open
Abstract
(1) Background: Little is known about the subsequent renal function change following incident infectious diseases in living-donor liver transplant (LT) recipients. (2) Methods: We studied patients who underwent living-donor LT from January 2003 to January 2019 to evaluate the association of incident hospitalization with major infections or pneumonia with adverse renal outcomes, including a sustained 40% reduction in estimated glomerular filtration rate (eGFR) and renal composite outcome (a 40% decline in eGFR, end-stage renal disease, or death.). Multivariable-adjusted time-dependent Cox models with infection as a time-varying exposure were used to estimate hazard ratio (HR) with 95% confidence interval (CI) for study outcomes. (3) Results: We identified 435 patients (mean age 54.6 ± 8.4 years and 76.3% men), of whom 102 had hospitalization with major infections during follow-up; the most common cause of infection was pneumonia (38.2%). In multiple Cox models, hospitalization with a major infection was associated with an increased risk of eGFR decline > 40% (HR, 3.32; 95% CI 2.13−5.16) and renal composite outcome (HR, 3.41; 95% CI 2.40−5.24). Likewise, pneumonia was also associated with an increased risk of eGFR decline > 40% (HR, 2.47; 95% CI 1.10−5.56) and renal composite outcome (HR, 4.37; 95% CI 2.39−8.02). (4) Conclusions: Our results illustrated the impact of a single infection episode on the future risk of adverse renal events in LT recipients. Whether preventive and prophylactic care bundles against infection and judicious modification of the immunosuppressive regimen benefit renal outcomes may deserve further study.
Collapse
|
34
|
Fu Y, Xiang Y, Li H, Chen A, Dong Z. Inflammation in kidney repair: Mechanism and therapeutic potential. Pharmacol Ther 2022; 237:108240. [PMID: 35803367 DOI: 10.1016/j.pharmthera.2022.108240] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 02/07/2023]
Abstract
The kidney has a remarkable ability of repair after acute kidney injury (AKI). However, when injury is severe or persistent, the repair is incomplete or maladaptive and may lead to chronic kidney disease (CKD). Maladaptive kidney repair involves multiple cell types and multifactorial processes, of which inflammation is a key component. In the process of inflammation, there is a bidirectional interplay between kidney parenchymal cells and the immune system. The extensive and complex crosstalk between renal tubular epithelial cells and interstitial cells, including immune cells, fibroblasts, and endothelial cells, governs the repair and recovery of the injured kidney. Further research in this field is imperative for the discovery of biomarkers and promising therapeutic targets for kidney repair. In this review, we summarize the latest progress in the immune response and inflammation during maladaptive kidney repair, analyzing the interaction between immune cells and intrinsic kidney cells, pointing out the potentialities of inflammation-related pathways as therapeutic targets, and discussing the challenges and future research prospects in this field.
Collapse
Affiliation(s)
- Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410011, China
| | - Yu Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410011, China
| | - Honglin Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410011, China
| | - Anqun Chen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410011, China
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410011, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
35
|
Xu L, Guo J, Moledina DG, Cantley LG. Immune-mediated tubule atrophy promotes acute kidney injury to chronic kidney disease transition. Nat Commun 2022; 13:4892. [PMID: 35986026 PMCID: PMC9391331 DOI: 10.1038/s41467-022-32634-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 08/05/2022] [Indexed: 01/12/2023] Open
Abstract
Incomplete repair after acute kidney injury can lead to development of chronic kidney disease. To define the mechanism of this response, we compared mice subjected to identical unilateral ischemia-reperfusion kidney injury with either contralateral nephrectomy (where tubule repair predominates) or contralateral kidney intact (where tubule atrophy predominates). By day 14, the kidneys undergoing atrophy had more macrophages with higher expression of chemokines, correlating with a second wave of proinflammatory neutrophil and T cell recruitment accompanied by increased expression of tubular injury genes and a decreased proportion of differentiated tubules. Depletion of neutrophils and T cells after day 5 reduced tubular cell loss and associated kidney atrophy. In kidney biopsies from patients with acute kidney injury, T cell and neutrophil numbers negatively correlated with recovery of estimated glomerular filtration rate. Together, our findings demonstrate that macrophage persistence after injury promotes a T cell- and neutrophil-mediated proinflammatory milieu and progressive tubule damage.
Collapse
Affiliation(s)
- Leyuan Xu
- Department of Internal Medicine/Section of Nephrology, Yale University School of Medicine, New Haven, CT, USA.
| | - Jiankan Guo
- Department of Internal Medicine/Section of Nephrology, Yale University School of Medicine, New Haven, CT, USA
| | - Dennis G Moledina
- Department of Internal Medicine/Section of Nephrology, Yale University School of Medicine, New Haven, CT, USA
| | - Lloyd G Cantley
- Department of Internal Medicine/Section of Nephrology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
36
|
DeWolf SE, Kasimsetty SG, Hawkes AA, Stocks LM, Kurian SM, McKay DB. DAMPs Released From Injured Renal Tubular Epithelial Cells Activate Innate Immune Signals in Healthy Renal Tubular Epithelial Cells. Transplantation 2022; 106:1589-1599. [PMID: 34954736 PMCID: PMC9218002 DOI: 10.1097/tp.0000000000004038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (IRI) predictably causes acute kidney injury after shock and major cardiovascular procedures in all kidneys procured for transplantation. The earliest events of IRI are triggered by molecules released from injured cells, damage-associated molecular patterns (DAMPs), that bind pattern recognition receptors (PRRs) constitutively expressed on many cells within the kidney. Activation of PRR signaling leads to production of proinflammatory molecules, which incite a cascade of inflammatory events leading to acute kidney injury. Renal tubular epithelial cells (RTECs) are particularly susceptible to ischemic injury, and proximal RTEC injury is pathognomonic of renal IRI. To better understand how injured RTECs contribute to the cycle of deleterious inflammation in the setting of renal IRI, this study asked whether DAMPs released from injured RTECs induced PRR signals in healthy RTECs. METHODS Human RTECs were necrosed ex vivo to release intracellular DAMPs and resulting necrotic supernatant used to stimulate healthy RTECs, T lymphocytes, and monocytes. RESULTS DAMPs released from necrosed RTECs upregulated PRRs known to be associated with renal IRI and activated mitogen-activated protein kinase signaling pathways. Proinflammatory cytokines were upregulated in response to necrotic supernatant, and this upregulation was abrogated by MEK-1 inhibition. The RTEC-derived DAMPs were also potent inducers of T-cell activation/proliferation and monocyte migration. CONCLUSIONS This is the first study to our knowledge to show that endogenous DAMPs released from injured RTECs directly activate PRR signaling in healthy RTECs. These findings provide new insights directed to therapeutics for renal IRI.
Collapse
Affiliation(s)
- Sean E DeWolf
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
- Department of Pulmonary and Critical Care Medicine, University of California San Diego, San Diego, CA
| | - Sashi G Kasimsetty
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
| | - Alana A Hawkes
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
| | - Lisa M Stocks
- LifeSharing Organ Procurement Organization, San Diego, CA
| | - Sunil M Kurian
- Division of Cell and Organ Transplantion, Scripps Clinic and Green Hospital, La Jolla, CA
| | - Dianne B McKay
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
- Division of Cell and Organ Transplantion, Scripps Clinic and Green Hospital, La Jolla, CA
| |
Collapse
|
37
|
Chen J, Luo P, Wang C, Yang C, Bai Y, He X, Zhang Q, Zhang J, Yang J, Wang S, Wang J. Integrated single-cell transcriptomics and proteomics reveal cellular-specific response and microenvironment remodeling in aristolochic acid nephropathy. JCI Insight 2022; 7:157360. [PMID: 35852860 PMCID: PMC9462482 DOI: 10.1172/jci.insight.157360] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Aristolochic acid nephropathy (AAN) is characterized by acute proximal tubule necrosis and immune cell infiltration, contributing to the global burden of chronic kidney disease and urothelial cancer. Although the proximal tubule has been defined as the primary target of aristolochic acids I (AAI), the mechanistic underpinning of gross renal deterioration caused by AAI has not been explicitly explained, prohibiting effective therapeutic intervention. To this point, we employed integrated single-cell RNA-Seq, bulk RNA-Seq, and mass spectrometry–based proteomics to analyze the mouse kidney after acute AAI exposure. Our results reveal a dramatic reduction of proximal tubule epithelial cells, associated with apoptotic and inflammatory pathways, indicating permanent damage beyond repair. We found the enriched development pathways in other nephron segments, suggesting activation of reparative programs triggered by AAI. The divergent response may be attributed to the segment-specific distribution of organic anion channels along the nephron, including OAT1 and OAT3. Moreover, we observed dramatic activation and recruitment of cytotoxic T and macrophage M1 cells, highlighting inflammation as a principal contributor to permanent renal injury. Ligand-receptor pairing revealed that critical intercellular crosstalk underpins damage-induced activation of immune cells. These results provide potentially novel insight into the AAI-induced kidney injury and point out possible pathways for future therapeutic intervention.
Collapse
Affiliation(s)
- Jiayun Chen
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Piao Luo
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chen Wang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yunmeng Bai
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xueling He
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Zhang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junzhe Zhang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Yang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Shuang Wang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Jigang Wang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Center for Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Southern Medical University, Dongguan, Guangdong, China
| |
Collapse
|
38
|
Sun D, Cui S, Ma H, Zhu P, Li N, Zhang X, Zhang L, Xuan L, Li J. Salvianolate ameliorates renal tubular injury through the Keap1/Nrf2/ARE pathway in mouse kidney ischemia-reperfusion injury. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115331. [PMID: 35489662 DOI: 10.1016/j.jep.2022.115331] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute kidney injury (AKI) is a common clinical disease characterized by rapid loss of renal function. Salvianolate is a prescribed Chinese medicine derived from traditional Chinese medicine Salvia miltiorrhiza bunge that possesses many pharmacological effects, the active components extracted from Salvia miltiorrhiza bunge have been proved to protect ischemia-reperfusion (I/R)-AKI. AIM OF THE STUDY This study aims to validate the therapeutic effect of SAL on I/R-AKI, and explore its potential pharmacological mechanism. MATERIALS AND METHODS Mice were pretreated with/without salvianolate (10, 30, and 90 mg/kg) before renal ischemia-reperfusion operation. Serum creatinine, BUN, and H&E staining were performed to evaluate renal function. Immunofluorescence analysis was conducted to measure renal tubular injury including inflammatory factors and peroxide level. Apoptosis of the kidney tissues was determined by TUNEL assay. Keap1-Nrf2-ARE and apoptosis signaling pathways were measured by Western blot, RT-PCR, and YO-PRO-1 staining in kidneys or NRK52E cells. RESULTS Pretreatment with SAL effectively alleviated renal function and ameliorated epithelial tubular injury, oxidative stress, and inflammatory response. Furthermore, the mechanistic study demonstrated that the SAL exerts anti-apoptotic effects through activation of the Keap1-Nrf2-ARE signaling pathway in renal tubular cells. CONCLUSION These findings indicate the therapeutic benefit of salvianolate in the protection of renal injury from ischemia-reperfusion, and strengthen the evidence for the AKI treatment strategy by the anti-oxidative stress response, suggesting that SAL may be a potential agent for the treatment of AKI.
Collapse
Affiliation(s)
- Dan Sun
- 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
| | - Shichao Cui
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haijian Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Pengfei Zhu
- The First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ni Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinwen Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lina Zhang
- Shanghai Green Valley Pharmaceutical Co.,Ltd, Shanghai, China
| | - Lijiang Xuan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jingya 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
|
39
|
Abstract
Sepsis-associated AKI is a life-threatening complication that is associated with high morbidity and mortality in patients who are critically ill. Although it is clear early supportive interventions in sepsis reduce mortality, it is less clear that they prevent or ameliorate sepsis-associated AKI. This is likely because specific mechanisms underlying AKI attributable to sepsis are not fully understood. Understanding these mechanisms will form the foundation for the development of strategies for early diagnosis and treatment of sepsis-associated AKI. Here, we summarize recent laboratory and clinical studies, focusing on critical factors in the pathophysiology of sepsis-associated AKI: microcirculatory dysfunction, inflammation, NOD-like receptor protein 3 inflammasome, microRNAs, extracellular vesicles, autophagy and efferocytosis, inflammatory reflex pathway, vitamin D, and metabolic reprogramming. Lastly, identifying these molecular targets and defining clinical subphenotypes will permit precision approaches in the prevention and treatment of sepsis-associated AKI.
Collapse
Affiliation(s)
- Shuhei Kuwabara
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Eibhlin Goggins
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
40
|
Aghsaeifard Z, Alizadeh R, Bagheri N. Association between neutrophil gelatinase-associated lipocalin (NGAL) and iron profile in chronic renal disease. Arch Physiol Biochem 2022; 128:703-707. [PMID: 31994917 DOI: 10.1080/13813455.2020.1720742] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
NGAL, also known as lipocalin 2, is a stress protein located on the cell surface that is known for its involvement in iron transport. This study is aimed to evaluate the correlation between the iron profile and NGAL concentration in serum among chronic kidney disease patients under dialysis in order to find its diagnostic value with regards to iron deficiency anaemia (IDA). 47 patients under chronic haemodialysis in end-stage renal disease (ESRD) and 15 healthy controls were evaluated to determine the correlation between serum NGAL concentration and IDA characteristics. Our results recorded a significant correlation between IDA (TSAT < 20%) and NGAL serum concentration with a Spearman's coefficient of 0.314. Serum NGAL was also significantly related to serum ferritin, TIBC, uric acid, creatinine and blood sugar whereas, an inverse relationship with albumin, total cholesterol and LDL. Our study reports a positive correlation between IDA and serum NGAL levels in CKD patients.
Collapse
Affiliation(s)
- Ziba Aghsaeifard
- Department of Internal Medicine, School of Medicine, Sina Hospital Tehran University of Medical Sciences, Tehran, Iran
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Alizadeh
- Department of Anesthesiology and Intensive Care, AJA University of Medical Sciences, Tehran, Iran
| | - Nazilla Bagheri
- Department of Adult Nephrology, School of Medicine, Ayatollah Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Xia W, Li C, Yao X, Chen Y, Zhang Y, Hu H. Prognostic value of fibrinogen to albumin ratios among critically ill patients with acute kidney injury. Intern Emerg Med 2022; 17:1023-1031. [PMID: 34850361 PMCID: PMC9135817 DOI: 10.1007/s11739-021-02898-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022]
Abstract
Fibrinogen to albumin ratios (FAR) have shown to be a promising prognostic factor for improving the predictive accuracy in various diseases. This study explores FAR's prognostic significance in critically ill patients with acute kidney injury (AKI). All clinical data were extracted from the Multiparameter Intelligent Monitoring in Intensive Care Database III version 1.4. All patients were divided into four groups based on FAR quartiles. The primary endpoint was in-hospital mortality. A generalized additive model was applied to explore a nonlinear association between FAR and in-hospital mortality. The Cox proportional hazards models were used to determine the association between FAR and in-hospital mortality. A total of 5001 eligible subjects were enrolled. Multivariate analysis demonstrated that higher FAR was an independent predictor of in-hospital mortality after adjusting for potential confounders (HR, 95% CI 1.23, 1.03-1.48, P = 0.025). A nonlinear relationship between FAR and in-hospital mortality was observed. FAR may serve as a potential prognostic biomarker in critically patients with AKI and higher FAR was associated with increased risk of in-hospital mortality among these patients.
Collapse
Affiliation(s)
- Wenkai Xia
- Department of Nephrology, The Affiliated Jiangyin Hospital of Southeast University Medical College, 3 Yinrui Road, Jiangsu, 214400, Jiangyin, China
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Chenyu Li
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Xiajuan Yao
- Department of Nephrology, The Affiliated Jiangyin Hospital of Southeast University Medical College, 3 Yinrui Road, Jiangsu, 214400, Jiangyin, China
| | - Yan Chen
- Department of Nephrology, The Affiliated Jiangyin Hospital of Southeast University Medical College, 3 Yinrui Road, Jiangsu, 214400, Jiangyin, China
| | - Yaoquan Zhang
- Department of Nephrology, The Affiliated Jiangyin Hospital of Southeast University Medical College, 3 Yinrui Road, Jiangsu, 214400, Jiangyin, China
| | - Hong Hu
- Department of Nephrology, The Affiliated Jiangyin Hospital of Southeast University Medical College, 3 Yinrui Road, Jiangsu, 214400, Jiangyin, China.
| |
Collapse
|
42
|
Tang W, Panja S, Jogdeo CM, Tang S, Ding L, Yu A, Foster KW, Dsouza DL, Chhonker YS, Jensen-Smith H, Jang HS, Boesen EI, Murry DJ, Padanilam B, Oupický D. Modified chitosan for effective renal delivery of siRNA to treat acute kidney injury. Biomaterials 2022; 285:121562. [PMID: 35552115 PMCID: PMC9133205 DOI: 10.1016/j.biomaterials.2022.121562] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/02/2022] [Accepted: 05/01/2022] [Indexed: 11/02/2022]
Abstract
Acute kidney injury (AKI) is characterized by a sudden decrease in renal function and impacts growing number of people worldwide. RNA interference (RNAi) showed potential to treat diseases with no or limited conventional therapies, including AKI. Suitable carriers are needed to protect and selectively deliver RNAi to target cells to fully explore this therapeutic modality. Here, we report on the synthesis of chitosan modified with α-cyclam-p-toluic acid (C-CS) as a novel siRNA carrier for targeted delivery to injured kidneys. We demonstrate that conjugation of the α-cyclam-p-toluic acid to chitosan imparts the C-CS polymer with targeting and antagonistic properties to cells overexpressing chemokine receptor CXCR4. In contrast, the parent α-cyclam-p-toluic acid showed no such properties. Self-assembled C-CS/siRNA nanoparticles rapidly accumulate in the injured kidneys and show long retention in renal tubules. Apoptosis and metabolic and inflammatory pathways induced by p53 are important pathological mechanisms in the development of AKI. Nanoparticles with siRNA against p53 (sip53) were formulated and intravenously injected for attenuation of IRI-AKI. Due to the favorable accumulation in injured kidneys, the treatment with C-CS/sip53 decreased renal injury, extent of renal apoptosis, macrophage and neutrophil infiltration, and improved renal function. Overall, our study suggests that C-CS/siRNA nanoparticles have the potential to effectively accumulate and deliver therapeutic siRNAs to injured kidneys through CXCR4 binding, providing a novel way for AKI therapy.
Collapse
Affiliation(s)
- Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ao Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kirk W Foster
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Del L Dsouza
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yashpal S Chhonker
- Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Heather Jensen-Smith
- Eppley Institute for Research in Cancer & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Hee-Seong Jang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Erika I Boesen
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Daryl J Murry
- Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Babu Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
43
|
Tang W, Panja S, Jogdeo CM, Tang S, Yu A, Oupický D. Study of Renal Accumulation of Targeted Polycations in Acute Kidney Injury. Biomacromolecules 2022; 23:2064-2074. [PMID: 35394757 PMCID: PMC9150723 DOI: 10.1021/acs.biomac.2c00079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Acute kidney injury (AKI) is a global healthcare burden characterized by rapid loss of renal function and high morbidity and mortality. Chemokine receptor CXCR4 participates in the renal infiltration of immune cells following injury and in local inflammatory enhancement. Injured renal tubule cells overexpress CXCR4, which could be used as a target for improved drug delivery in AKI. Plerixafor is a small-molecule CXCR4 antagonist that has shown beneficial effects against AKI and has been previously developed into a polymeric analog (polymeric plerixafor, PP). With the goal of gaining a better understanding of how overall charge and hydrophilicity affect renal accumulation of PP, we have synthesized PP copolymers containing hydroxyl, carboxyl, primary amine, and alkyl moieties using Michael-type addition copolymerization. All synthesized copolymers showed excellent CXCR4-binding and inhibiting ability in vitro and improved cellular uptake in hypoxia-reoxygenation stimulated mouse tubule cells. Analysis of serum protein binding revealed that polymers with hydroxyl group modification showed the least amount of protein binding. Biodistribution of the polymers was tested in a unilateral ischemia reperfusion-induced AKI mouse model. The results showed significant differences in accumulation in the injured kidneys depending on the net charge and hydrophilicity of the polymers. The findings of this study will guide the development of polymeric drug carriers for targeted delivery to injured kidneys for better AKI therapy.
Collapse
Affiliation(s)
- Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chinmay M. Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ao Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
44
|
Aal-Aaboda M, Abu Raghif AR, Almudhafer RH, Hadi NR. Lipopolysaccharide from Rhodobacter spheroids modulate toll-like receptors expression and tissue damage in an animal model of bilateral renal ischemic reperfusion injury. J Med Life 2022; 15:685-697. [PMID: 35815074 PMCID: PMC9262262 DOI: 10.25122/jml-2021-0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022] Open
Abstract
Ischemic reperfusion injury (IRI) of the kidneys is a direct sequela of surgical procedures associated with the interruption of blood supply. The pathophysiology of IRI is complicated, and several inflammatories, apoptosis, and oxidative stress pathways are implicated. Among the major receptors directly involved in renal IRI are the toll-like receptors (TLRs), specifically TLR2 and TLR4. In this study, we investigated the effects of Lipopolysaccharide from Rhodobacter Sphaeroides (TLR2 and TLR4 antagonist, LPS-RS) and the ultrapure form (pure TLR4 antagonist, ULPS-RS) on the histopathological changes and TLRs expression in an animal model of bilateral renal IRI. Forty-eight adult male rats were allocated into six groups (N=8) as follows: sham group (negative control without IRI), control group (rats underwent bilateral renal ischemia for 30 minutes and 2 hours of reperfusion), vehicle group (IRI+ vehicle), LPS-RS group (IRI+ 0.5 mg/kg of LPS-RS), ULPS-RS group (IRI+ 0.1 mg/kg of ULPS-RS), ULPS-RSH group (IRI+ 0.2 mg/kg of ULPS-RS). Significant improvement in the histopathological damages induced by renal IRI was found in the ULPS-RS treated groups at both doses compared with the control group. The protective effect of ULPS-RS was associated with significantly reduced TLR4 expression without affecting TLR2. Regarding LPS-RS, the tested dose adversely affected the renal tissues as manifested by the histopathological findings, although it similarly affected TLRs expression as ULPS-RS. Our results demonstrated that ULPS-RS was renoprotective while LPS-RS had no protective effect against the tissue damages induced by renal IRI.
Collapse
Affiliation(s)
- Munaf Aal-Aaboda
- Department of Pharmacology, Faculty of Pharmacy, University of Misan, Amarah, Iraq
| | | | - Rihab Hameed Almudhafer
- Middle Euphrates Unit for Cancer Research, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Najah Riesh Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq,Corresponding Author: Najah Riesh Hadi, Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq. E-mail:
| |
Collapse
|
45
|
Sun W, Choi HS, Kim CS, Bae EH, Ma SK, Kim SW. Maslinic Acid Attenuates Ischemia/Reperfusion-Induced Acute Kidney Injury by Suppressing Inflammation and Apoptosis Through Inhibiting NF-κB and MAPK Signaling Pathway. Front Pharmacol 2022; 13:807452. [PMID: 35496304 PMCID: PMC9039024 DOI: 10.3389/fphar.2022.807452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/03/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation and apoptosis are the major contributors to the mechanisms of acute kidney injury (AKI) due to renal ischemia-reperfusion injury (IRI). Maslinic acid (MA), a pentacyclic triterpene acid mostly found in dietary plants, the current study was to demonstrate the renoprotective effect of MA on IRI-induced AKI, and to investigate the role of inflammation and apoptosis-related signaling pathways as a molecular mechanism. C57BL/6J mice were subjected to IRI for 72 h, and MA was daily administered by intraperitoneal injection during this period. In parallel, rat renal proximal tubule cells (NRK52E) were prophylactically treated with MA and then exposed to hydrogen peroxide (H2O2). MA treatment significantly inhibited the mRNA expression of interleukin (IL-1β), tumor necrosis factor-α (TGF-α), monocyte chemoattractant protein-1 (MCP-1), and intercellular adhesion molecule-1(ICAM-1). Also, MA reduced the expression of Bax/Bcl2 ratio and cleaved caspase-3. In NRK52 cells, MA inhibited the IκBα degradation, blocked NF-κB/p65 phosphorylation, and nuclear translocation. The phosphorylation of ERK, JNK, and p38 was attenuated by MA in IRI-induced kidney injury and H2O2-stimulated NRK52 cells. The expression levels of IL-1β, MCP-1, and ICAM-1 were upregulated in H2O2-stimulated NRK52E cells, which was attenuated by NF-κB inhibitor. H2O2 treatment increased the Bax/Bcl2 ratio and cleaved caspase-3 in NRK52E cells, which was counteracted by MAPK inhibitors. Together, our data demonstrate that MA suppresses IR-induced AKI injury through NF-κB and MAPK signaling pathways and that MA is a promising agent in the treatment of kidney diseases.
Collapse
|
46
|
Jiang H, Li D, Xu T, Chen Z, Shan Y, Zhao L, Fu G, Luan Y, Xia S, Zhang W. Systemic Immune-Inflammation Index Predicts Contrast-Induced Acute Kidney Injury in Patients Undergoing Coronary Angiography: A Cross-Sectional Study. Front Med (Lausanne) 2022; 9:841601. [PMID: 35372392 PMCID: PMC8965764 DOI: 10.3389/fmed.2022.841601] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/16/2022] [Indexed: 01/21/2023] Open
Abstract
Background and Aims Systemic immune-inflammation index (SII) is an emerging indicator and correlated to the incidence of cardiovascular diseases. This study aimed to explore the association between SII and contrast-induced acute kidney injury (CI-AKI). Methods In this retrospective cross-sectional study, 4,381 subjects undergoing coronary angiography (CAG) were included. SII is defined as neutrophil count × platelet count/lymphocyte count. CI-AKI was determined by the elevation of serum creatinine (Scr). Multivariable linear and logistic regression analysis were used to determine the relationship of SII with Scr and CI-AKI, respectively. Receiver operator characteristic (ROC) analysis, structural equation model analysis, and subgroup analysis were also performed. Results Overall, 786 (17.9%) patients suffered CI-AKI after the intravascular contrast administration. The subjects were 67.1 ± 10.8 years wold, with a mean SII of 5.72 × 1011/L. Multivariable linear regression analysis showed that SII linearly increased with the proportion of Scr elevation (β [95% confidence interval, CI] = 0.315 [0.206 to 0.424], P < 0.001). Multivariable logistic regression analysis demonstrated that higher SII was associated with an increased incidence of CI-AKI ([≥12 vs. <3 × 1011/L]: odds ratio, OR [95% CI] = 2.914 [2.121 to 4.003], P < 0.001). Subgroup analysis showed consistent results. ROC analysis identified a good predictive value of SII on CI-AKI (area under the ROC curve [95% CI]: 0.625 [0.602 to 0.647]). The structural equation model verified a more remarkable direct effect of SII (β = 0.102, P < 0.001) on CI-AKI compared to C-reactive protein (β = 0.070, P < 0.001). Conclusions SII is an independent predictor for CI-AKI in patients undergoing CAG procedures.
Collapse
Affiliation(s)
- Hangpan Jiang
- Department of Cardiology, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu, China
| | - Duanbin Li
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Tian Xu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Zhezhe Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Yu Shan
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Liding Zhao
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Shudong Xia
- Department of Cardiology, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu, China
- *Correspondence: Shudong Xia
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
- Wenbin Zhang
| |
Collapse
|
47
|
Hematological Ratios Are Associated with Acute Kidney Injury and Mortality in Patients That Present with Suspected Infection at the Emergency Department. J Clin Med 2022; 11:jcm11041017. [PMID: 35207289 PMCID: PMC8874958 DOI: 10.3390/jcm11041017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/03/2022] [Accepted: 02/10/2022] [Indexed: 01/27/2023] Open
Abstract
The early recognition of acute kidney injury (AKI) is essential to improve outcomes and prevent complications such as chronic kidney disease, the need for renal-replacement therapy, and an increased length of hospital stay. Increasing evidence shows that inflammation plays an important role in the pathophysiology of AKI and mortality. Several inflammatory hematological ratios can be used to measure systemic inflammation. Therefore, the association between these ratios and outcomes (AKI and mortality) in patients suspected of having an infection at the emergency department was investigated. Data from the SPACE cohort were used. Cox regression was performed to investigate the association between seven hematological ratios and outcomes. A total of 1889 patients were included, of which 160 (8.5%) patients developed AKI and 102 (5.4%) died in <30 days. The Cox proportional-hazards model revealed that the neutrophil-to-lymphocyte ratio (NLR), segmented-neutrophil-to-monocyte ratio (SMR), and neutrophil-lymphocyte-platelet ratio (NLPR) are independently associated with AKI <30 days after emergency-department presentation. Additionally, the NLR, SMR and NLPR were associated with 30-day all-cause mortality. These findings are an important step forward for the early recognition of AKI. The use of these markers might enable emergency-department physicians to recognize and treat AKI in an early phase to potentially prevent complications.
Collapse
|
48
|
Templeton EM, Lassé M, Kleffmann T, Ellmers LJ, Palmer SC, Davidson T, Scott NJA, Pickering JW, Charles CJ, Endre ZH, Cameron VA, Richards AM, Rademaker MT, Pilbrow AP. Identifying Candidate Protein Markers of Acute Kidney Injury in Acute Decompensated Heart Failure. Int J Mol Sci 2022; 23:ijms23021009. [PMID: 35055195 PMCID: PMC8778509 DOI: 10.3390/ijms23021009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/15/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022] Open
Abstract
One-quarter of patients with acute decompensated heart failure (ADHF) experience acute kidney injury (AKI)—an abrupt reduction or loss of kidney function associated with increased long-term mortality. There is a critical need to identify early and real-time markers of AKI in ADHF; however, to date, no protein biomarkers have exhibited sufficient diagnostic or prognostic performance for widespread clinical uptake. We aimed to identify novel protein biomarkers of AKI associated with ADHF by quantifying changes in protein abundance in the kidneys that occur during ADHF development and recovery in an ovine model. Relative quantitative protein profiling was performed using sequential window acquisition of all theoretical fragment ion spectra–mass spectrometry (SWATH–MS) in kidney cortices from control sheep (n = 5), sheep with established rapid-pacing-induced ADHF (n = 8), and sheep after ~4 weeks recovery from ADHF (n = 7). Of the 790 proteins quantified, we identified 17 candidate kidney injury markers in ADHF, 1 potential kidney marker of ADHF recovery, and 2 potential markers of long-term renal impairment (differential abundance between groups of 1.2–2.6-fold, adjusted p < 0.05). Among these 20 candidate protein markers of kidney injury were 6 candidates supported by existing evidence and 14 novel candidates not previously implicated in AKI. Proteins of differential abundance were enriched in pro-inflammatory signalling pathways: glycoprotein VI (activated during ADHF development; adjusted p < 0.01) and acute phase response (repressed during recovery from ADHF; adjusted p < 0.01). New biomarkers for the early detection of AKI in ADHF may help us to evaluate effective treatment strategies to prevent mortality and improve outcomes for patients.
Collapse
Affiliation(s)
- Evelyn M. Templeton
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
- Correspondence: ; Tel.: +64-03-364-12-53
| | - Moritz Lassé
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Torsten Kleffmann
- Research Infrastructure Centre, Division of Health Sciences, University of Otago, Dunedin 9016, New Zealand;
| | - Leigh J. Ellmers
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Suetonia C. Palmer
- Department of Medicine, University of Otago, Christchurch 8014, New Zealand;
| | - Trent Davidson
- Department of Anatomical Pathology, Prince of Wales Hospital, Sydney, NSW 2031, Australia;
| | - Nicola J. A. Scott
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - John W. Pickering
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Christopher J. Charles
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Zoltan H. Endre
- Department of Nephrology, Prince of Wales Hospital, Sydney, NSW 2031, Australia;
| | - Vicky A. Cameron
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - A. Mark Richards
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
- Cardiovascular Research Institute, Department of Cardiology, National University of Singapore, Singapore 119077, Singapore
| | - Miriam T. Rademaker
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| | - Anna P. Pilbrow
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand; (M.L.); (L.J.E.); (N.J.A.S.); (J.W.P.); (C.J.C.); (V.A.C.); (A.M.R.); (M.T.R.); (A.P.P.)
| |
Collapse
|
49
|
Rojas-Morales P, León-Contreras JC, Sánchez-Tapia M, Silva-Palacios A, Cano-Martínez A, González-Reyes S, Jiménez-Osorio AS, Hernández-Pando R, Osorio-Alonso H, Sánchez-Lozada LG, Tovar AR, Pedraza-Chaverri J, Tapia E. A ketogenic diet attenuates acute and chronic ischemic kidney injury and reduces markers of oxidative stress and inflammation. Life Sci 2022; 289:120227. [PMID: 34921866 DOI: 10.1016/j.lfs.2021.120227] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/30/2021] [Accepted: 12/08/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND Ischemic kidney injury is a common clinical condition resulting from transient interruption of the kidney's normal blood flow, leading to oxidative stress, inflammation, and kidney dysfunction. The ketogenic diet (KD), a low-carbohydrate, high-fat diet that stimulates endogenous ketone body production, has potent antioxidant and anti-inflammatory effects in distinct tissues and might thus protect the kidney against ischemia and reperfusion (IR) injury. MAIN METHODS Male Wistar rats were fed a KD or a control diet (CD) for three days before analyzing metabolic parameters or testing nephroprotection. We used two different models of kidney IR injury and conducted biochemical, histological, and Western blot analyses at 24 h and two weeks after surgery. KEY FINDINGS Acute KD feeding caused protein acetylation, liver AMPK activation, and increased resistance to IR-induced kidney injury. At 24 h after IR, rats on KD presented reduced tubular damage and improved kidney functioning compared to rats fed with a CD. KD attenuated oxidative damage (protein nitration, 4-HNE adducts, and 8-OHdG), increased antioxidant defenses (GPx and SOD activity), and reduced inflammatory intermediates (IL6, TNFα, MCP1), p50 NF-κB expression, and cellular infiltration. Also, KD prevented interstitial fibrosis development at two weeks, up-regulation of HSP70, and chronic Klotho deficiency. SIGNIFICANCE Our findings demonstrate for the first time that short-term KD increases tolerance to experimental kidney ischemia, opening the opportunity for future therapeutic exploration of a dietary preconditioning strategy to convey kidney protection in the clinic.
Collapse
Affiliation(s)
- Pedro Rojas-Morales
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico; Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Juan Carlos León-Contreras
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Mónica Sánchez-Tapia
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Susana González-Reyes
- Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Tijuana 22390, Mexico
| | - Angélica Saraí Jiménez-Osorio
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda, La Concepción S/N, Carretera Pachuca Actopan, San Agustín Tlaxiaca 42060, Hidalgo, Mexico
| | - Rogelio Hernández-Pando
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Laura Gabriela Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Edilia Tapia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico.
| |
Collapse
|
50
|
McBride WT, Kurth MJ, Domanska A, Watt J, McLean G, Joseph J, Lamont JV, Fitzgerald P, Ruddock MW. Blood and urinary cytokine balance and renal outcomes at cardiac surgery. BMC Nephrol 2021; 22:406. [PMID: 34876054 PMCID: PMC8653550 DOI: 10.1186/s12882-021-02621-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Increased perioperative pro-inflammatory biomarkers, renal hypoperfusion and ischemia reperfusion injury (IRI) heighten cardiac surgery acute kidney injury (CS-AKI) risk. Increased urinary anti-inflammatory cytokines attenuate risk. We evaluated whether blood and urinary anti-inflammatory biomarkers, when expressed as ratios with biomarkers of inflammation, hypoperfusion and IRI are increased in CS-AKI patients. METHODS Preoperative and 24-h postoperative blood and urinary pro-inflammatory and anti-inflammatory cytokines, blood VEGF and H-FABP (hypoperfusion biomarkers), and MK, a biomarker for IRI, were measured in 401 cardiac surgery patients. Pre- and postoperative concentrations of biomarkers and selected ratios thereof, were compared between non-CS-AKI and CS-AKI patients. RESULTS Compared with non-CS-AKI, blood pro-inflammatory (pre- and post-op TNFα, IP-10, IL-12p40, MIP-1α, NGAL; pre-op IL-6; post-op IL-8, MK) and anti-inflammatory (pre- and post-op sTNFsr1, sTNFsr2, IL-1RA) biomarkers together with urinary pro-inflammatory (pre- and post-op uIL-12p40; post-op uIP-10, uNGAL) and anti-inflammatory (pre- and post-op usTNFsr1, usTNFsr2, uIL-1RA) biomarkers, were significantly higher in CS-AKI patients. Urinary anti-inflammatory biomarkers, when expressed as ratios with biomarkers of inflammation (blood and urine), hypoperfusion (blood H-FABP and VEGF) and IRI (blood MK) were decreased in CS-AKI. In contrast, blood anti-inflammatory biomarkers expressed as similar ratios with blood biomarkers were increased in CS-AKI. CONCLUSIONS The urinary anti-inflammatory response may protect against the injurious effects of perioperative inflammation, hypoperfusion and IRI. These finding may have clinical utility in bioprediction and earlier diagnosis of CS-AKI and informing future therapeutic strategies for CS-AKI patients.
Collapse
Affiliation(s)
- William T. McBride
- Department of Cardiac Anesthesia, Belfast Health & Social Care Trust, 274 Grosvenor Road, Belfast, Northern Ireland BT12 6BA UK
| | - Mary Jo Kurth
- Randox Laboratories Ltd, Clinical Studies Group, 55 Diamond Road, Crumlin, County Antrim BT29 4QY Northern Ireland, UK
| | - Anna Domanska
- Randox Laboratories Ltd, Clinical Studies Group, 55 Diamond Road, Crumlin, County Antrim BT29 4QY Northern Ireland, UK
| | - Joanne Watt
- Randox Laboratories Ltd, Clinical Studies Group, 55 Diamond Road, Crumlin, County Antrim BT29 4QY Northern Ireland, UK
| | - Gavin McLean
- Department of Cardiac Anesthesia, Belfast Health & Social Care Trust, 274 Grosvenor Road, Belfast, Northern Ireland BT12 6BA UK
| | - Jijin Joseph
- Department of Cardiac Anesthesia, Belfast Health & Social Care Trust, 274 Grosvenor Road, Belfast, Northern Ireland BT12 6BA UK
| | - John V. Lamont
- Randox Laboratories Ltd, Clinical Studies Group, 55 Diamond Road, Crumlin, County Antrim BT29 4QY Northern Ireland, UK
| | - Peter Fitzgerald
- Randox Laboratories Ltd, Clinical Studies Group, 55 Diamond Road, Crumlin, County Antrim BT29 4QY Northern Ireland, UK
| | - Mark W. Ruddock
- Randox Laboratories Ltd, Clinical Studies Group, 55 Diamond Road, Crumlin, County Antrim BT29 4QY Northern Ireland, UK
| |
Collapse
|