1
|
Hao S, Wang H, Li S, Zhang H, Xie X, Liu J, Yang C, Zhou W, Wang H. Carbon monoxide polyhemoglobin improves the therapeutic effect and relieves inflammation in the colon tissue of haemorrhagic shock/resuscitation rats. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:370-383. [PMID: 39017642 DOI: 10.1080/21691401.2024.2367444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/03/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVE The objective of this study was to test the therapeutic effect of carbon monoxide polyhemoglobin (polyCOHb) in haemorrhagic shock/resuscitation and its underlying mechanisms. METHODS 48 rats were divided into two experimental parts, and 36 rats in the first experiment and 12 rats in the second experiment. In the first experimental part, 36 animals were randomly assigned to the following groups: hydroxyethyl starch group (HES group, n = 12), polyhemoglobin group (polyHb group, n = 12), and carbon monoxide polyhemoglobin group (polyCOHb group, n = 12). In the second experimental part, 12 animals were randomly assigned to the following groups: polyHb group (n = 6), and polyCOHb group (n = 6). Then the anaesthetised rats were haemorrhaged by withdrawing 50% of the animal's blood volume (BV), and resuscitated to the same volume of the animal's withdrawing BV with HES, polyHb, polyCOHb. In the first experimental part, the 72h survival rates of each groups animals were measured. In the second experimental part, the rats' mean arterial pressure (MAP), heart rate (HR), blood gas levels and other indicators were dynamically monitored in baseline, haemorrhagic shock (HS), at 0point resuscitation (RS 0h) and after 1 h resuscitation (RS 1h). The concentrations of malondialdehyde (MDA), superoxide dismutase (SOD), tumour necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) were measured by ELISA kits in both groups of rats at RS 1h. Changes in pathological sections were examined by haematoxylin-eosin (HE) staining. Nuclear factor erythroid 2-related factor 2 (Nrf2) and haem oxygenase-1 (HO-1) levels were detected by immunohistochemical analysis, while myeloperoxidase (MPO) levels were detected by immunofluorescence. DHE staining was used to determine reactive oxygen species (ROS) levels. RESULTS The 72h survival rates of the polyHb and polyCOHb groups were 50.00% (6/12) and 58.33% (7/12) respectively, which were significantly higher than that of the 8.33% (1/12) in the HES group (p < 0.05). At RS 0h and RS 1h, the HbCO content of rats in the polyCOHb group (1.90 ± 0.21, 0.80 ± 0.21) g/L were higher than those in the polyHb group (0.40 ± 0.09, 0.50 ± 0.12)g/L (p < 0.05); At RS 1h, the MDA (41.47 ± 3.89 vs 34.17 ± 3.87 nmol/ml) in the plasma, Nrf2 and HO-1 content in the colon of rats in the polyCOHb group were lower than the polyHb group. And the SOD in the plasma (605.01 ± 24.46 vs 678.64 ± 36.37) U/mg and colon (115.72 ± 21.17 vs 156.70 ± 21.34) U/mg and the MPO content in the colon in the polyCOHb group were higher than the polyHb group (p < 0.05). CONCLUSIONS In these haemorrhagic shock/resuscitation models, both polyCOHb and polyHb show similar therapeutic effects, and polyCOHb has more effective effects in maintaining MAP, correcting acidosis, reducing inflammatory responses than that in polyHb.
Collapse
Affiliation(s)
- Shasha Hao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Huan Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Shen Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Honghui Zhang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Xintong Xie
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Jiaxin Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Chengmin Yang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Wentao Zhou
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Hong Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
2
|
Ghajar-Rahimi G, Barwinska D, Whipple GE, Kamocka MM, Khan S, Winfree S, Lafontaine J, Soliman RH, Melkonian AL, Zmijewska AA, Cheung MD, Traylor AM, Jiang Y, Yang Z, Bolisetty S, Zarjou A, Lee T, George JF, El-Achkar TM, Agarwal A. Acute kidney injury results in long-term alterations of kidney lymphatics in mice. Am J Physiol Renal Physiol 2024; 327:F869-F884. [PMID: 39323387 DOI: 10.1152/ajprenal.00120.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/14/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024] Open
Abstract
The long-term effects of a single episode of acute kidney injury (AKI) induced by bilateral ischemia-reperfusion injury (BIRI) on kidney lymphatic dynamics are not known. The purpose of this study was to determine if alterations in kidney lymphatics are sustained in the long term and how they relate to inflammation and injury. Mice underwent BIRI as a model of AKI and were followed up to 9 mo. Although kidney function markers normalized following initial injury, histological analysis revealed sustained tissue damage and inflammation for up to 9 mo. Transcriptional analysis showed both acute and late-stage lymphangiogenesis, supported by increased expression of lymphatic markers, with unique signatures at each phase. Expression of Ccl21a was distinctly upregulated during late-stage lymphangiogenesis. Three-dimensional tissue cytometry confirmed increased lymphatic vessel abundance, particularly in the renal cortex, at early and late timepoints postinjury. In addition, the study identified the formation of tertiary lymphoid structures composed of CCR7+ lymphocytes and observed changes in immune cell composition over time, suggesting a complex and dynamic response to AKI involving tissue remodeling and immune cell involvement. This study provides new insights into the role of lymphatics in the progression of AKI to chronic kidney disease.NEW & NOTEWORTHY Here, we perform the first, comprehensive study of long-term lymphatic dynamics following a single acute kidney injury (AKI) event. Using improved three-dimensional image analysis and an expanded panel of transcriptional markers, we identify multiple stages of lymphatic responses with distinct transcriptional signatures, associations with the immune microenvironment, and collagen deposition. This research advances kidney lymphatic biology, emphasizing the significance of longitudinal studies in understanding AKI and the transition to chronic kidney disease.
Collapse
Affiliation(s)
- Gelare Ghajar-Rahimi
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Daria Barwinska
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Grace E Whipple
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Malgorzata M Kamocka
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Shehnaz Khan
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Seth Winfree
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Jennifer Lafontaine
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama, United States
| | - Reham H Soliman
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Arin L Melkonian
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Anna A Zmijewska
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Matthew D Cheung
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Amie M Traylor
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Yanlin Jiang
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Zhengqin Yang
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Subhashini Bolisetty
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Abolfazl Zarjou
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Timmy Lee
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama, United States
| | - James F George
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tarek M El-Achkar
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Indianapolis Veterans Affairs Medical Center, Indianapolis, Indiana, United States
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
3
|
Yuan Y, Li H, Sreeram K, Malankhanova T, Boddu R, Strader S, Chang A, Bryant N, Yacoubian TA, Standaert DG, Erb M, Moore DJ, Sanders LH, Lutz MW, Velmeshev D, West AB. Single molecule array measures of LRRK2 kinase activity in serum link Parkinson's disease severity to peripheral inflammation. Mol Neurodegener 2024; 19:47. [PMID: 38862989 PMCID: PMC11167795 DOI: 10.1186/s13024-024-00738-4] [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: 02/22/2024] [Accepted: 06/02/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND LRRK2-targeting therapeutics that inhibit LRRK2 kinase activity have advanced to clinical trials in idiopathic Parkinson's disease (iPD). LRRK2 phosphorylates Rab10 on endolysosomes in phagocytic cells to promote some types of immunological responses. The identification of factors that regulate LRRK2-mediated Rab10 phosphorylation in iPD, and whether phosphorylated-Rab10 levels change in different disease states, or with disease progression, may provide insights into the role of Rab10 phosphorylation in iPD and help guide therapeutic strategies targeting this pathway. METHODS Capitalizing on past work demonstrating LRRK2 and phosphorylated-Rab10 interact on vesicles that can shed into biofluids, we developed and validated a high-throughput single-molecule array assay to measure extracellular pT73-Rab10. Ratios of pT73-Rab10 to total Rab10 measured in biobanked serum samples were compared between informative groups of transgenic mice, rats, and a deeply phenotyped cohort of iPD cases and controls. Multivariable and weighted correlation network analyses were used to identify genetic, transcriptomic, clinical, and demographic variables that predict the extracellular pT73-Rab10 to total Rab10 ratio. RESULTS pT73-Rab10 is absent in serum from Lrrk2 knockout mice but elevated by LRRK2 and VPS35 mutations, as well as SNCA expression. Bone-marrow transplantation experiments in mice show that serum pT73-Rab10 levels derive primarily from circulating immune cells. The extracellular ratio of pT73-Rab10 to total Rab10 is dynamic, increasing with inflammation and rapidly decreasing with LRRK2 kinase inhibition. The ratio of pT73-Rab10 to total Rab10 is elevated in iPD patients with greater motor dysfunction, irrespective of disease duration, age, sex, or the usage of PD-related or anti-inflammatory medications. pT73-Rab10 to total Rab10 ratios are associated with neutrophil degranulation, antigenic responses, and suppressed platelet activation. CONCLUSIONS The extracellular serum ratio of pT73-Rab10 to total Rab10 is a novel pharmacodynamic biomarker for LRRK2-linked innate immune activation associated with disease severity in iPD. We propose that those iPD patients with higher serum pT73-Rab10 levels may benefit from LRRK2-targeting therapeutics that mitigate associated deleterious immunological responses.
Collapse
Affiliation(s)
- Yuan Yuan
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Huizhong Li
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Kashyap Sreeram
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Tuyana Malankhanova
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Ravindra Boddu
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Samuel Strader
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Allison Chang
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Nicole Bryant
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Talene A Yacoubian
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David G Standaert
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Madalynn Erb
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Laurie H Sanders
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA
- Department of Neurology, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
| | - Michael W Lutz
- Department of Neurology, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
| | | | - Andrew B West
- Duke Center for Neurodegeneration and Neurotheraputics, Duke University, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Neurology, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
4
|
Yuan Y, Li H, Sreeram K, Malankhanova T, Boddu R, Strader S, Chang A, Bryant N, Yacoubian TA, Standaert DG, Erb M, Moore DJ, Sanders LH, Lutz MW, Velmeshev D, West AB. Single molecule array measures of LRRK2 kinase activity in serum link Parkinson's disease severity to peripheral inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589570. [PMID: 38659797 PMCID: PMC11042295 DOI: 10.1101/2024.04.15.589570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Background LRRK2-targeting therapeutics that inhibit LRRK2 kinase activity have advanced to clinical trials in idiopathic Parkinson's disease (iPD). LRRK2 phosphorylates Rab10 on endolysosomes in phagocytic cells to promote some types of immunological responses. The identification of factors that regulate LRRK2-mediated Rab10 phosphorylation in iPD, and whether phosphorylated-Rab10 levels change in different disease states, or with disease progression, may provide insights into the role of Rab10 phosphorylation in iPD and help guide therapeutic strategies targeting this pathway. Methods Capitalizing on past work demonstrating LRRK2 and phosphorylated-Rab10 interact on vesicles that can shed into biofluids, we developed and validated a high-throughput single-molecule array assay to measure extracellular pT73-Rab10. Ratios of pT73-Rab10 to total Rab10 measured in biobanked serum samples were compared between informative groups of transgenic mice, rats, and a deeply phenotyped cohort of iPD cases and controls. Multivariable and weighted correlation network analyses were used to identify genetic, transcriptomic, clinical, and demographic variables that predict the extracellular pT73-Rab10 to total Rab10 ratio. Results pT73-Rab10 is absent in serum from Lrrk2 knockout mice but elevated by LRRK2 and VPS35 mutations, as well as SNCA expression. Bone-marrow transplantation experiments in mice show that serum pT73-Rab10 levels derive primarily from circulating immune cells. The extracellular ratio of pT73-Rab10 to total Rab10 is dynamic, increasing with inflammation and rapidly decreasing with LRRK2 kinase inhibition. The ratio of pT73-Rab10 to total Rab10 is elevated in iPD patients with greater motor dysfunction, irrespective of disease duration, age, sex, or the usage of PD-related or anti-inflammatory medications. pT73-Rab10 to total Rab10 ratios are associated with neutrophil activation, antigenic responses, and the suppression of platelet activation. Conclusions The extracellular ratio of pT73-Rab10 to total Rab10 in serum is a novel pharmacodynamic biomarker for LRRK2-linked innate immune activation associated with disease severity in iPD. We propose that those iPD patients with higher serum pT73-Rab10 levels may benefit from LRRK2-targeting therapeutics to mitigate associated deleterious immunological responses.
Collapse
|
5
|
Athanassiadou V, Plavoukou S, Grapsa E, Detsika MG. The Role of Heme Oxygenase-1 as an Immunomodulator in Kidney Disease. Antioxidants (Basel) 2022; 11:antiox11122454. [PMID: 36552662 PMCID: PMC9774641 DOI: 10.3390/antiox11122454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
The protein heme oxygenase (HO)-1 has been implicated in the regulations of multiple immunological processes. It is well known that kidney injury is affected by immune mechanisms and that various kidney-disease forms may be a result of autoimmune disease. The current study describes in detail the role of HO-1 in kidney disease and provides the most recent observations of the effect of HO-1 on immune pathways and responses both in animal models of immune-mediated disease forms and in patient studies.
Collapse
Affiliation(s)
- Virginia Athanassiadou
- Department of Nephrology, School of Medicine, National and Kapodistrian University of Athens, Aretaieion University Hospital, 11528 Athens, Greece
| | - Stella Plavoukou
- Department of Nephrology, School of Medicine, National and Kapodistrian University of Athens, Aretaieion University Hospital, 11528 Athens, Greece
| | - Eirini Grapsa
- Department of Nephrology, School of Medicine, National and Kapodistrian University of Athens, Aretaieion University Hospital, 11528 Athens, Greece
| | - Maria G. Detsika
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, 10675 Athens, Greece
- Correspondence:
| |
Collapse
|
6
|
Ghajar-Rahimi G, Traylor AM, Mathew B, Bostwick JR, Nebane NM, Zmijewska AA, Esman SK, Thukral S, Zhai L, Sambandam V, Cowell RM, Suto MJ, George JF, Augelli-Szafran CE, Agarwal A. Identification of Cytoprotective Small-Molecule Inducers of Heme-Oxygenase-1. Antioxidants (Basel) 2022; 11:1888. [PMID: 36290611 PMCID: PMC9598442 DOI: 10.3390/antiox11101888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Acute kidney injury (AKI) is a major public health concern with significant morbidity and mortality and no current treatments beyond supportive care and dialysis. Preclinical studies have suggested that heme-oxygenase-1 (HO-1), an enzyme that catalyzes the breakdown of heme, has promise as a potential therapeutic target for AKI. Clinical trials involving HO-1 products (biliverdin, carbon monoxide, and iron), however, have not progressed beyond the Phase ½ level. We identified small-molecule inducers of HO-1 that enable us to exploit the full therapeutic potential of HO-1, the combination of its products, and yet-undefined effects of the enzyme system. Through cell-based, high-throughput screens for induction of HO-1 driven by the human HO-1 promoter/enhancer, we identified two novel small molecules and broxaldine (an FDA-approved drug) for further consideration as candidate compounds exhibiting an Emax ≥70% of 5 µM hemin and EC50 <10 µM. RNA sequencing identified shared binding motifs to NRF2, a transcription factor known to regulate antioxidant genes, including HMOX1. In vitro, the cytoprotective function of the candidates was assessed against cisplatin-induced cytotoxicity and apoptosis. In vivo, delivery of a candidate compound induced HO-1 expression in the kidneys of mice. This study serves as the basis for further development of small-molecule HO-1 inducers as preventative or therapeutic interventions for a variety of pathologies, including AKI.
Collapse
Affiliation(s)
- Gelare Ghajar-Rahimi
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Amie M. Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Bini Mathew
- Southern Research, Birmingham, AL 35205, USA
| | | | | | - Anna A. Zmijewska
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Stephanie K. Esman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Saakshi Thukral
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ling Zhai
- Southern Research, Birmingham, AL 35205, USA
| | | | - Rita M. Cowell
- Southern Research, Birmingham, AL 35205, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | - James F. George
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Veterans Affairs, Birmingham, AL 35233, USA
| |
Collapse
|
7
|
Different Acute Kidney Injury Patterns after Renal Ischemia Reperfusion Injury and Extracorporeal Membrane Oxygenation in Mice. Int J Mol Sci 2022; 23:ijms231911000. [PMID: 36232304 PMCID: PMC9570202 DOI: 10.3390/ijms231911000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
The use of extracorporeal membrane oxygenation (ECMO) is associated with acute kidney injury (AKI) in thoracic organ transplantation. However, multiple other factors contribute to AKI development after these procedures such as renal ischemia-reperfusion injury (IRI) due to hypo-perfusion of the kidney during surgery. In this study, we aimed to explore the kidney injury patterns in mouse models of ECMO and renal IRI. Kidneys of C57BL/6 mice were examined after moderate (35 min) and severe (45 min) unilateral transient renal pedicle clamping and 2 h of veno-venous ECMO. Renal injury markers, neutrophil infiltration, tubular transport function, pro-inflammatory cytokines, and renal heme oxygenase-1 (HO-1) expression were determined by immunofluorescence and qPCR. Both procedures caused AKI, but with different injury patterns. Severe neutrophil infiltration of the kidney was evident after renal IRI, but not following ECMO. Tubular transport function was severely impaired after renal IRI, but preserved in the ECMO group. Both procedures caused upregulation of pro-inflammatory cytokines in the renal tissue, but with different time kinetics. After ECMO, but not IRI, HO-1 was strongly induced in tubular cells indicating contact with hemolysis-derived proteins. After IRI, HO-1 was expressed on infiltrating myeloid cells in the tubulo-interstitial space. In conclusion, renal IRI and ECMO both caused AKI, but kidney damage after renal IRI was more pronounced including severe neutrophil infiltration and tubular transport impairment. Enhanced HO-1 expression in tubular cells after ECMO encourages limitation of hemolysis as a therapeutic approach to reduce ECMO-associated AKI.
Collapse
|
8
|
Kwong AM, Luke PPW, Bhattacharjee RN. Carbon monoxide mechanism of protection against renal ischemia and reperfusion injury. Biochem Pharmacol 2022; 202:115156. [PMID: 35777450 DOI: 10.1016/j.bcp.2022.115156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/20/2022]
Abstract
Carbon monoxide is quickly moving past its historic label as a molecule once feared, to a therapeutic drug that modulates inflammation. The development of carbon monoxide releasing molecules and utilization of heme oxygenase-1 inducers have shown carbon monoxide to be a promising therapy in reducing renal ischemia and reperfusion injury and other inflammatory diseases. In this review, we will discuss the developments and application of carbon monoxide releasing molecules in renal ischemia and reperfusion injury, and transplantation. We will review the anti-inflammatory mechanisms of carbon monoxide in respect to mitigating apoptosis, suppressing dendritic cell maturation and signalling, inhibiting toll-like receptor activation, promoting anti-inflammatory responses, and the effects on renal vasculature.
Collapse
Affiliation(s)
- Aaron M Kwong
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Patrick P W Luke
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Surgery, London Health Sciences Centre, Canada; Matthew Mailing Centre for Translational Transplantation Studies, Canada.
| | - Rabindra N Bhattacharjee
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Surgery, London Health Sciences Centre, Canada; Matthew Mailing Centre for Translational Transplantation Studies, Canada.
| |
Collapse
|
9
|
Cheung MD, Agarwal A, George JF. Where Are They Now: Spatial and Molecular Diversity of Tissue-Resident Macrophages in the Kidney. Semin Nephrol 2022; 42:151276. [PMID: 36435683 DOI: 10.1016/j.semnephrol.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Kidney resident macrophages (KRMs) are involved in homeostasis, phagocytosis, defense against infectious agents, response to insults, inflammation, and tissue repair. They also play critical roles in the pathogenesis and recovery from many kidney diseases such as acute kidney injury. KRMs historically have been studied as one homogenous population, but the wide-ranging roles and phenotypes observed suggest that there is greater heterogeneity than previously understood. Advancements in RNA sequencing technologies (single-cell RNA sequencing and spatial transcriptomics) have identified specific subsets of KRMs that are molecularly, functionally, and spatially distinct with dynamic changes after kidney injury. Multiple studies have identified unique markers that represent these subpopulations, permitting further characterization of the function and roles they play in the kidney. Understanding the diversity of KRM subpopulations will be key in the development of novel therapies used in treating kidney diseases and promoting kidney health.
Collapse
Affiliation(s)
- Matthew D Cheung
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Department of Veteran Affairs, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - James F George
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama.
| |
Collapse
|
10
|
Li Y, Ma K, Han Z, Chi M, Sai X, Zhu P, Ding Z, Song L, Liu C. Immunomodulatory Effects of Heme Oxygenase-1 in Kidney Disease. Front Med (Lausanne) 2021; 8:708453. [PMID: 34504854 PMCID: PMC8421649 DOI: 10.3389/fmed.2021.708453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/31/2021] [Indexed: 01/23/2023] Open
Abstract
Kidney disease is a general term for heterogeneous damage that affects the function and the structure of the kidneys. The rising incidence of kidney diseases represents a considerable burden on the healthcare system, so the development of new drugs and the identification of novel therapeutic targets are urgently needed. The pathophysiology of kidney diseases is complex and involves multiple processes, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Heme oxygenase-1 (HO-1), an enzyme involved in the process of heme degradation, has attracted widespread attention in recent years due to its cytoprotective properties. As an enzyme with known anti-oxidative functions, HO-1 plays an indispensable role in the regulation of oxidative stress and is involved in the pathogenesis of several kidney diseases. Moreover, current studies have revealed that HO-1 can affect cell proliferation, cell maturation, and other metabolic processes, thereby altering the function of immune cells. Many strategies, such as the administration of HO-1-overexpressing macrophages, use of phytochemicals, and carbon monoxide-based therapies, have been developed to target HO-1 in a variety of nephropathological animal models, indicating that HO-1 is a promising protein for the treatment of kidney diseases. Here, we briefly review the effects of HO-1 induction on specific immune cell populations with the aim of exploring the potential therapeutic roles of HO-1 and designing HO-1-based therapeutic strategies for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Yunlong Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Zhongyu Han
- School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingxuan Chi
- School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyalatu Sai
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhaolun Ding
- Department of Emergency Surgery, Shannxi Provincial People's Hospital, Xi'an, China
| | - Linjiang Song
- School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
De La Cruz LK, Yang X, Menshikh A, Brewer M, Lu W, Wang M, Wang S, Ji X, Cachuela A, Yang H, Gallo D, Tan C, Otterbein L, de Caestecker M, Wang B. Adapting decarbonylation chemistry for the development of prodrugs capable of in vivo delivery of carbon monoxide utilizing sweeteners as carrier molecules. Chem Sci 2021; 12:10649-10654. [PMID: 34447558 PMCID: PMC8356820 DOI: 10.1039/d1sc02711e] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Carbon monoxide as an endogenous signaling molecule exhibits pharmacological efficacy in various animal models of organ injury. To address the difficulty in using CO gas as a therapeutic agent for widespread applications, we are interested in developing CO prodrugs through bioreversible caging of CO in an organic compound. Specifically, we have explored the decarboxylation-decarbonylation chemistry of 1,2-dicarbonyl compounds. Examination and optimization of factors favorable for maximal CO release under physiological conditions led to organic CO prodrugs using non-calorific sweeteners as leaving groups attached to the 1,2-dicarbonyl core. Attaching a leaving group with appropriate properties promotes the desired hydrolysis-decarboxylation-decarbonylation sequence of reactions that leads to CO generation. One such CO prodrug was selected to recapitulate the anti-inflammatory effects of CO against LPS-induced TNF-α production in cell culture studies. Oral administration in mice elevated COHb levels to the safe and efficacious levels established in various preclinical and clinical studies. Furthermore, its pharmacological efficacy was demonstrated in mouse models of acute kidney injury. These studies demonstrate the potential of these prodrugs with benign carriers as orally active CO-based therapeutics. This represents the very first example of orally active organic CO prodrugs with a benign carrier that is an FDA-approved sweetener with demonstrated safety profiles in vivo.
Collapse
Affiliation(s)
| | - Xiaoxiao Yang
- Department of Chemistry, Georgia State University Atlanta GA 30303 USA
| | - Anna Menshikh
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - Maya Brewer
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - Wen Lu
- Department of Chemistry, Georgia State University Atlanta GA 30303 USA
| | - Minjia Wang
- Department of Pharmaceutics and Drug Delivery, University of Mississippi MS 38677 USA
| | - Siming Wang
- Department of Chemistry, Georgia State University Atlanta GA 30303 USA
| | - Xingyue Ji
- Department of Chemistry, Georgia State University Atlanta GA 30303 USA
| | - Alyssa Cachuela
- Department of Chemistry, Georgia State University Atlanta GA 30303 USA
| | - Haichun Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - David Gallo
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School Boston MA 02115 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi MS 38677 USA
| | - Leo Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School Boston MA 02115 USA
| | - Mark de Caestecker
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - Binghe Wang
- Department of Chemistry, Georgia State University Atlanta GA 30303 USA
| |
Collapse
|
12
|
Mertens C, Marques O, Horvat NK, Simonetti M, Muckenthaler MU, Jung M. The Macrophage Iron Signature in Health and Disease. Int J Mol Sci 2021; 22:ijms22168457. [PMID: 34445160 PMCID: PMC8395084 DOI: 10.3390/ijms22168457] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Throughout life, macrophages are located in every tissue of the body, where their main roles are to phagocytose cellular debris and recycle aging red blood cells. In the tissue niche, they promote homeostasis through trophic, regulatory, and repair functions by responding to internal and external stimuli. This in turn polarizes macrophages into a broad spectrum of functional activation states, also reflected in their iron-regulated gene profile. The fast adaptation to the environment in which they are located helps to maintain tissue homeostasis under physiological conditions.
Collapse
Affiliation(s)
- Christina Mertens
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Correspondence: (C.M.); (M.J.); Tel.: +(49)-622-156-4582 (C.M.); +(49)-696-301-6931 (M.J.)
| | - Oriana Marques
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
| | - Natalie K. Horvat
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Collaboration for Joint PhD Degree between EMBL and the Faculty of Biosciences, University of Heidelberg, 69117 Heidelberg, Germany
| | - Manuela Simonetti
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, INF 366, 69120 Heidelberg, Germany;
| | - Martina U. Muckenthaler
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; (O.M.); (N.K.H.); (M.U.M.)
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Correspondence: (C.M.); (M.J.); Tel.: +(49)-622-156-4582 (C.M.); +(49)-696-301-6931 (M.J.)
| |
Collapse
|
13
|
Matsuura R, Yamashita T, Hayase N, Hamasaki Y, Noiri E, Numata G, Takimoto E, Nangaku M, Doi K. Preexisting heart failure with reduced ejection fraction attenuates renal fibrosis after ischemia reperfusion via sympathetic activation. Sci Rep 2021; 11:15091. [PMID: 34302012 PMCID: PMC8302613 DOI: 10.1038/s41598-021-94617-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 07/05/2021] [Indexed: 11/17/2022] Open
Abstract
Although chronic heart failure is clinically associated with acute kidney injury (AKI), the precise mechanism that connects kidney and heart remains unknown. Here, we elucidate the effect of pre-existing heart failure with reduced ejection fraction (HFrEF) on kidney via sympathetic activity, using the combining models of transverse aortic constriction (TAC) and unilateral renal ischemia reperfusion (IR). The evaluation of acute (24 h) and chronic (2 weeks) phases of renal injury following IR 8 weeks after TAC in C57BL/6 mice revealed that the development of renal fibrosis in chronic phase was significantly attenuated in TAC mice, but not in non-TAC mice, whereas no impact of pre-existing heart failure was observed in acute phase of renal IR. Expression of transforming growth factor-β, monocyte chemoattractant protein-1, and macrophage infiltration were significantly reduced in TAC mice. Lastly, to investigate the effect of sympathetic nerve activity, we performed renal sympathetic denervation two days prior to renal IR, which abrogated attenuation of renal fibrosis in TAC mice. Collectively, we demonstrate the protective effect of pre-existing HFrEF on long-term renal ischemic injury. Renal sympathetic nerve may contribute to this protection; however, further studies are needed to fully clarify the comprehensive mechanisms associated with attenuated renal fibrosis and pre-existing HFrEF.
Collapse
Affiliation(s)
- Ryo Matsuura
- Department of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsushi Yamashita
- Department of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoki Hayase
- Department of Acute Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655, Japan
| | - Yoshifumi Hamasaki
- Department of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eisei Noiri
- Department of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Genri Numata
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaomi Nangaku
- Department of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kent Doi
- Department of Acute Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655, Japan.
| |
Collapse
|
14
|
Nash WT, Okusa MD. Chess Not Checkers: Complexities Within the Myeloid Response to the Acute Kidney Injury Syndrome. Front Med (Lausanne) 2021; 8:676688. [PMID: 34124107 PMCID: PMC8187556 DOI: 10.3389/fmed.2021.676688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/26/2021] [Indexed: 12/23/2022] Open
Abstract
Immune dysregulation in acute kidney injury (AKI) is an area of intense interest which promises to enhance our understanding of the disease and how to manage it. Macrophages are a heterogeneous and dynamic population of immune cells that carry out multiple functions in tissue, ranging from maintenance to inflammation. As key sentinels of their environment and the major immune population in the uninjured kidney, macrophages are poised to play an important role in the establishment and pathogenesis of AKI. These cells have a profound capacity to orchestrate downstream immune responses and likely participate in skewing the kidney environment toward either pathogenic inflammation or injury resolution. A clear understanding of macrophage and myeloid cell dynamics in the development of AKI will provide valuable insight into disease pathogenesis and options for intervention. This review considers evidence in the literature that speaks to the role and regulation of macrophages and myeloid cells in AKI. We also highlight barriers or knowledge gaps that need to be addressed as the field advances.
Collapse
Affiliation(s)
- William T Nash
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, United States
| | - Mark D Okusa
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
15
|
Curtis LM, George J, Vallon V, Barnes S, Darley-Usmar V, Vaingankar S, Cutter GR, Gutierrez OM, Seifert M, Ix JH, Mehta RL, Sanders PW, Agarwal A. UAB-UCSD O'Brien Center for Acute Kidney Injury Research. Am J Physiol Renal Physiol 2021; 320:F870-F882. [PMID: 33779316 DOI: 10.1152/ajprenal.00661.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Acute kidney injury (AKI) remains a significant clinical problem through its diverse etiologies, the challenges of robust measurements of injury and recovery, and its progression to chronic kidney disease (CKD). Bridging the gap in our knowledge of this disorder requires bringing together not only the technical resources for research but also the investigators currently endeavoring to expand our knowledge and those who might bring novel ideas and expertise to this important challenge. The University of Alabama at Birmingham-University of California-San Diego O'Brien Center for Acute Kidney Injury Research brings together technical expertise and programmatic and educational efforts to advance our knowledge in these diverse issues and the required infrastructure to develop areas of novel exploration. Since its inception in 2008, this O'Brien Center has grown its impact by providing state-of-the-art resources in clinical and preclinical modeling of AKI, a bioanalytical core that facilitates measurement of critical biomarkers, including serum creatinine via LC-MS/MS among others, and a biostatistical resource that assists from design to analysis. Through these core resources and with additional educational efforts, our center has grown its investigator base to include >200 members from 51 institutions. Importantly, this center has translated its pilot and catalyst funding program with a $37 return per dollar invested. Over 500 publications have resulted from the support provided with a relative citation ratio of 2.18 ± 0.12 (iCite). Through its efforts, this disease-centric O'Brien Center is providing the infrastructure and focus to help the development of the next generation of researchers in the basic and clinical science of AKI. This center creates the promise of the application at the bedside of the advances in AKI made by current and future investigators.
Collapse
Affiliation(s)
- Lisa M Curtis
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - James George
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Volker Vallon
- Division of Nephrology, Department of Medicine, University of California-San Diego, San Diego, California
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sucheta Vaingankar
- Division of Pediatric Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gary R Cutter
- School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Orlando M Gutierrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael Seifert
- Division of Pediatric Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joachim H Ix
- Division of Nephrology, Department of Medicine, University of California-San Diego, San Diego, California
| | - Ravindra L Mehta
- Division of Nephrology, Department of Medicine, University of California-San Diego, San Diego, California
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Veterans Affairs, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Veterans Affairs, Birmingham, Alabama
| |
Collapse
|
16
|
Rossi M, Korpak K, Doerfler A, Zouaoui Boudjeltia K. Deciphering the Role of Heme Oxygenase-1 (HO-1) Expressing Macrophages in Renal Ischemia-Reperfusion Injury. Biomedicines 2021; 9:biomedicines9030306. [PMID: 33809696 PMCID: PMC8002311 DOI: 10.3390/biomedicines9030306] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 12/30/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a leading cause of acute kidney injury (AKI), which contributes to the development of chronic kidney disease (CKD). Renal IRI combines major events, including a strong inflammatory immune response leading to extensive cell injuries, necrosis and late interstitial fibrosis. Macrophages act as key players in IRI-induced AKI by polarizing into proinflammatory M1 and anti-inflammatory M2 phenotypes. Compelling evidence exists that the stress-responsive enzyme, heme oxygenase-1 (HO-1), mediates protection against renal IRI and modulates macrophage polarization by enhancing a M2 subset. Hereafter, we review the dual effect of macrophages in the pathogenesis of IRI-induced AKI and discuss the critical role of HO-1 expressing macrophages.
Collapse
Affiliation(s)
- Maxime Rossi
- Department of Urology, CHU de Charleroi, Université libre de Bruxelles (ULB), 6000 Charleroi, Belgium;
- Laboratory of Experimental Medicine (ULB 222 Unit), CHU de Charleroi, Hôpital André Vésale, Université libre de Bruxelles (ULB), 6110 Montigny-le-Tilleul, Belgium;
- Correspondence: (M.R.); (K.Z.B.)
| | - Kéziah Korpak
- Laboratory of Experimental Medicine (ULB 222 Unit), CHU de Charleroi, Hôpital André Vésale, Université libre de Bruxelles (ULB), 6110 Montigny-le-Tilleul, Belgium;
- Department of Geriatric Medicine, CHU de Charleroi, Hôpital André Vésale, Université libre de Bruxelles (ULB), 6110 Montigny-le-Tilleul, Belgium
| | - Arnaud Doerfler
- Department of Urology, CHU de Charleroi, Université libre de Bruxelles (ULB), 6000 Charleroi, Belgium;
| | - Karim Zouaoui Boudjeltia
- Laboratory of Experimental Medicine (ULB 222 Unit), CHU de Charleroi, Hôpital André Vésale, Université libre de Bruxelles (ULB), 6110 Montigny-le-Tilleul, Belgium;
- Correspondence: (M.R.); (K.Z.B.)
| |
Collapse
|
17
|
Grunenwald A, Roumenina LT, Frimat M. Heme Oxygenase 1: A Defensive Mediator in Kidney Diseases. Int J Mol Sci 2021; 22:2009. [PMID: 33670516 PMCID: PMC7923026 DOI: 10.3390/ijms22042009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
The incidence of kidney disease is rising, constituting a significant burden on the healthcare system and making identification of new therapeutic targets increasingly urgent. The heme oxygenase (HO) system performs an important function in the regulation of oxidative stress and inflammation and, via these mechanisms, is thought to play a role in the prevention of non-specific injuries following acute renal failure or resulting from chronic kidney disease. The expression of HO-1 is strongly inducible by a wide range of stimuli in the kidney, consequent to the kidney's filtration role which means HO-1 is exposed to a wide range of endogenous and exogenous molecules, and it has been shown to be protective in a variety of nephropathological animal models. Interestingly, the positive effect of HO-1 occurs in both hemolysis- and rhabdomyolysis-dominated diseases, where the kidney is extensively exposed to heme (a major HO-1 inducer), as well as in non-heme-dependent diseases such as hypertension, diabetic nephropathy or progression to end-stage renal disease. This highlights the complexity of HO-1's functions, which is also illustrated by the fact that, despite the abundance of preclinical data, no drug targeting HO-1 has so far been translated into clinical use. The objective of this review is to assess current knowledge relating HO-1's role in the kidney and its potential interest as a nephroprotection agent. The potential therapeutic openings will be presented, in particular through the identification of clinical trials targeting this enzyme or its products.
Collapse
Affiliation(s)
- Anne Grunenwald
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Lubka T. Roumenina
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Marie Frimat
- U1167-RID-AGE, Institut Pasteur de Lille, Inserm, Univ. Lille, F-59000 Lille, France
- Nephrology Department, CHU Lille, Univ. Lille, F-59000 Lille, France
| |
Collapse
|
18
|
Detsika MG, Lianos EA. Regulation of Complement Activation by Heme Oxygenase-1 (HO-1) in Kidney Injury. Antioxidants (Basel) 2021; 10:antiox10010060. [PMID: 33418934 PMCID: PMC7825075 DOI: 10.3390/antiox10010060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/31/2020] [Accepted: 01/01/2021] [Indexed: 12/20/2022] Open
Abstract
Heme oxygenase is a cytoprotective enzyme with strong antioxidant and anti-apoptotic properties. Its cytoprotective role is mainly attributed to its enzymatic activity, which involves the degradation of heme to biliverdin with simultaneous release of carbon monoxide (CO). Recent studies uncovered a new cytoprotective role for heme oxygenase-1 (HO-1) by identifying a regulatory role on the complement control protein decay-accelerating factor. This is a key complement regulatory protein preventing dysregulation or overactivation of complement cascades that can cause kidney injury. Cell-specific targeting of HO-1 induction may, therefore, be a novel approach to attenuate complement-dependent forms of kidney disease.
Collapse
Affiliation(s)
- Maria G. Detsika
- First Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M. Simou Laboratories, National & Kapodistrian University of Athens, Medical School, Evangelismos Hospital, 10675 Athens, Greece
- Correspondence: ; Tel.: +30-210-723552; Fax: +30-210-7239127
| | - Elias A. Lianos
- Thorax Foundation, Research Center of Intensive Care and Emergency Thoracic Medicine, 10675 Athens, Greece;
- Veterans Affairs Medical Center and Virginia Tech, Carilion School of Medicine, 1970 Roanoke Blvd, Salem, VA 24153, USA
| |
Collapse
|
19
|
Nath M, Agarwal A. New insights into the role of heme oxygenase-1 in acute kidney injury. Kidney Res Clin Pract 2020; 39:387-401. [PMID: 33184238 PMCID: PMC7770992 DOI: 10.23876/j.krcp.20.091] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022] Open
Abstract
Acute kidney injury (AKI) is attended by injury-related biomarkers appearing in the urine and serum, decreased urine output, and impaired glomerular filtration rate. AKI causes increased morbidity and mortality and can progress to chronic kidney disease and end-stage kidney failure. AKI is without specific therapies and is managed by supported care. Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. HO-1 induction protects against assorted renal insults as demonstrated by in vitro and preclinical models. This review summarizes the advances in understanding of the protection conferred by HO-1 in AKI, how HO-1 can be induced including via its transcription factor Nrf2, and HO-1 induction as a therapeutic strategy.
Collapse
Affiliation(s)
- Meryl Nath
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Veterans Affairs, Birmingham Veterans Administration Medical Center, Birmingham, AL, USA
| |
Collapse
|
20
|
Rossi M, Delbauve S, Roumeguère T, Wespes E, Leo O, Flamand V, Le Moine A, Hougardy JM. HO-1 mitigates acute kidney injury and subsequent kidney-lung cross-talk. Free Radic Res 2019; 53:1035-1043. [PMID: 31530210 DOI: 10.1080/10715762.2019.1668936] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ischemia-reperfusion injury (IRI) is a leading cause of acute kidney injury (AKI), which contributes to the development of chronic kidney disease (CKD). IRI-induced AKI releases proinflammatory cytokines (e.g. IL-1β, TNF-α, IL-6) that induce a systemic inflammatory response, resulting in proinflammatory cells recruitment and remote organ damage. AKI is associated with poor outcomes, particularly when extrarenal complications or distant organ injuries occur. Acute lung injury (ALI) is a major remote organ dysfunction associated with AKI. Hence, kidney-lung cross-talk remains a clinical challenge, especially in critically ill population. The stress-responsive enzyme, heme oxygenase-1 (HO-1) is largely known to protect against renal IRI and may be preventively induced using hemin prior to renal insult. However, the use of hemin-induced HO-1 to prevent AKI-induced ALI remains poorly investigated. Mice received an intraperitoneal injection of hemin or sterile saline 1 day prior to surgery. Twenty-four hours later, mice underwent bilateral renal IRI for 26 min or sham surgery. After 4 or 24 h of reperfusion, mice were sacrificed. Hemin-induced HO-1 improved renal outcomes after IRI (i.e. fewer renal damage, renal inflammation, and oxidative stress). This protective effect was associated with a dampened systemic inflammation (i.e. IL-6 and KC). Subsequently, mitigated lung inflammation was found in hemin-treated mice (i.e. neutrophils influx and lung KC). The present study demonstrates that hemin-induced HO-1 controls the magnitude of renal IRI and the subsequent AKI-induced ALI. Therefore, targeting HO-1 represents a promising approach to prevent the impact of renal IRI on distant organs, such as lung.
Collapse
Affiliation(s)
- Maxime Rossi
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Gosselies , Belgium.,Department of Urology, CUB Hôpital Erasme, Université Libre de Bruxelles , Brussels , Belgium.,Department of Urology, CHU-Charleroi, Université Libre de Bruxelles , Charleroi , Belgium
| | - Sandrine Delbauve
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Gosselies , Belgium
| | - Thierry Roumeguère
- Department of Urology, CUB Hôpital Erasme, Université Libre de Bruxelles , Brussels , Belgium
| | - Eric Wespes
- Department of Urology, CHU-Charleroi, Université Libre de Bruxelles , Charleroi , Belgium
| | - Oberdan Leo
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Gosselies , Belgium.,Laboratory of Immunobiology, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles , Gosselies , Belgium
| | - Véronique Flamand
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Gosselies , Belgium
| | - Alain Le Moine
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Gosselies , Belgium.,Department of Nephrology, Dialysis and Renal Transplantation, CUB Hôpital Erasme, Université Libre de Bruxelles , Brussels , Belgium
| | - Jean-Michel Hougardy
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Gosselies , Belgium.,Department of Nephrology, Dialysis and Renal Transplantation, CUB Hôpital Erasme, Université Libre de Bruxelles , Brussels , Belgium
| |
Collapse
|
21
|
Zarjou A, Black LM, Bolisetty S, Traylor AM, Bowhay SA, Zhang MZ, Harris RC, Agarwal A. Dynamic signature of lymphangiogenesis during acute kidney injury and chronic kidney disease. J Transl Med 2019; 99:1376-1388. [PMID: 31019289 PMCID: PMC6716993 DOI: 10.1038/s41374-019-0259-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/07/2019] [Accepted: 03/29/2019] [Indexed: 11/09/2022] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected syndromes with significant attributable morbidity and mortality. The disturbing trend of increasing incidence and prevalence of these clinical disorders highlights the urgent need for better understanding of the underlying mechanisms that are involved in pathogenesis of these conditions. Lymphangiogenesis and its involvement in various inflammatory conditions is increasingly recognized while its role in AKI and CKD remains to be fully elucidated. Here, we studied lymphangiogenesis in three models of kidney injury. Our results demonstrate that the main ligands for lymphangiogenesis, VEGF-C and VEGF-D, are abundantly present in tubules at baseline conditions and the expression pattern of these ligands is significantly altered following injury. In addition, we show that both of these ligands increase in serum and urine post-injury and suggest that such increment may serve as novel urinary biomarkers of AKI as well as in progression of kidney disease. We also provide evidence that irrespective of the nature of initial insult, lymphangiogenic pathways are rapidly and robustly induced as evidenced by higher expression of lymphatic markers within the kidney.
Collapse
Affiliation(s)
- Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Laurence M Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sarah A Bowhay
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ming-Zhi Zhang
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, USA
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Nashville Veterans Affairs Hospital, Nashville, TN, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA.
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Veterans Affairs, Birmingham, AL, USA.
| |
Collapse
|
22
|
Black LM, Lever JM, Agarwal A. Renal Inflammation and Fibrosis: A Double-edged Sword. J Histochem Cytochem 2019; 67:663-681. [PMID: 31116067 PMCID: PMC6713973 DOI: 10.1369/0022155419852932] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/30/2019] [Indexed: 12/29/2022] Open
Abstract
Renal tissue injury initiates inflammatory and fibrotic processes that occur to promote regeneration and repair. After renal injury, damaged tissue releases cytokines and chemokines, which stimulate activation and infiltration of inflammatory cells to the kidney. Normal tissue repair processes occur simultaneously with activation of myofibroblasts, collagen deposition, and wound healing responses; however, prolonged activation of pro-inflammatory and pro-fibrotic cell types causes excess extracellular matrix deposition. This review focuses on the physiological and pathophysiological roles of specialized cell types, cytokines/chemokines, and growth factors, and their implications in recovery or exacerbation of acute kidney injury.
Collapse
Affiliation(s)
- Laurence M Black
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Jeremie M Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Anupam Agarwal
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL
- Department of Veterans Affairs, The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
23
|
Abstract
Iron is required for key aspects of cellular physiology including mitochondrial function and DNA synthesis and repair. However, free iron is an aberration because of its ability to donate electrons, reduce oxygen, and generate reactive oxygen species. Iron-mediated cell injury or ferroptosis is a central player in the pathogenesis of acute kidney injury. There are several homeostatic proteins and pathways that maintain critical balance in iron homeostasis to allow iron's biologic functions yet avoid ferroptosis. Hepcidin serves as the master regulator of iron homeostasis through its ability to regulate ferroportin-mediated iron export and intracellular H-ferritin levels. Hepcidin is a protective molecule in acute kidney injury. Drugs targeting hepcidin, H-ferritin, and ferroptosis pathways hold great promise to prevent or treat kidney injury. In this review we discuss iron homeostasis under physiological and pathologic conditions and highlight its importance in acute kidney injury.
Collapse
|
24
|
Pretreatment with Cholecalciferol Alleviates Renal Cellular Stress Response during Ischemia/Reperfusion-Induced Acute Kidney Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1897316. [PMID: 31019650 PMCID: PMC6452543 DOI: 10.1155/2019/1897316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/06/2018] [Accepted: 02/18/2019] [Indexed: 02/06/2023]
Abstract
Background Cellular stress is involved in ischemia/reperfusion- (I/R-) induced acute kidney injury (AKI). This study is aimed at investigating the effects of pretreatment with cholecalciferol on renal oxidative stress and endoplasmic reticulum (ER) stress during I/R-induced AKI. Methods I/R-induced AKI was established by cross-clamping renal pedicles for 90 minutes and then reperfusion. In the Chol + I/R group, mice were orally administered with three doses of cholecalciferol (25 μg/kg) at 1, 24, and 48 h before ischemia. Renal cellular stress and kidney injury were measured at different time points after reperfusion. Results I/R-induced AKI was alleviated in mice pretreated with cholecalciferol. In addition, I/R-induced renal cell apoptosis, as determined by TUNEL, was suppressed by cholecalciferol. Additional experiment showed that I/R-induced upregulation of renal GRP78 and CHOP was inhibited by cholecalciferol. I/R-induced renal IRE1α and eIF2α phosphorylation was attenuated by cholecalciferol. Moreover, I/R-induced renal GSH depletion, lipid peroxidation, and protein nitration were blocked in mice pretreated with cholecalciferol. I/R-induced upregulation of renal NADPH oxidases, such as p47phox, gp91phox, and nox4, was inhibited by cholecalciferol. I/R-induced upregulation of heme oxygenase- (HO-) 1, gshpx and gshrd, was attenuated in mice pretreated with cholecalciferol. Conclusions Pretreatment with cholecalciferol protects against I/R-induced AKI partially through suppressing renal cellular stress response.
Collapse
|
25
|
Chen T, Cao Q, Wang Y, Harris DCH. M2 macrophages in kidney disease: biology, therapies, and perspectives. Kidney Int 2019; 95:760-773. [PMID: 30827512 DOI: 10.1016/j.kint.2018.10.041] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/10/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022]
Abstract
Tissue macrophages are crucial players in homeostasis, inflammation, and immunity. They are characterized by heterogeneity and plasticity, due to which they display a continuum of phenotypes with M1/M2 presenting 2 extremes of this continuum. M2 macrophages are usually termed in the literature as anti-inflammatory and wound healing. Substantial progress has been made in elucidating the biology of M2 macrophages and their potential for clinical translation. In this review we discuss the current state of knowledge in M2 macrophage research with an emphasis on kidney disease. We explore their therapeutic potential and the challenges in using them as cellular therapies. Some new regulators that shape macrophage polarization and potential areas for future research are also examined.
Collapse
Affiliation(s)
- Titi Chen
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia; Center for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia.
| | - Qi Cao
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia; Center for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Yiping Wang
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia; Center for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - David C H Harris
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia; Center for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia
| |
Collapse
|
26
|
Zarjou A, Black LM, McCullough KR, Hull TD, Esman SK, Boddu R, Varambally S, Chandrashekar DS, Feng W, Arosio P, Poli M, Balla J, Bolisetty S. Ferritin Light Chain Confers Protection Against Sepsis-Induced Inflammation and Organ Injury. Front Immunol 2019; 10:131. [PMID: 30804939 PMCID: PMC6371952 DOI: 10.3389/fimmu.2019.00131] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/16/2019] [Indexed: 12/31/2022] Open
Abstract
Despite the prevalence and recognition of its detrimental impact, clinical complications of sepsis remain a major challenge. Here, we investigated the effects of myeloid ferritin heavy chain (FtH) in regulating the pathogenic sequelae of sepsis. We demonstrate that deletion of myeloid FtH leads to protection against lipopolysaccharide-induced endotoxemia and cecal ligation and puncture (CLP)-induced model of sepsis as evidenced by reduced cytokine levels, multi-organ dysfunction and mortality. We identified that such protection is predominantly mediated by the compensatory increase in circulating ferritin (ferritin light chain; FtL) in the absence of myeloid FtH. Our in vitro and in vivo studies indicate that prior exposure to ferritin light chain restrains an otherwise dysregulated response to infection. These findings are mediated by an inhibitory action of FtL on NF-κB activation, a key signaling pathway that is implicated in the pathogenesis of sepsis. We further identified that LPS mediated activation of MAPK pathways, specifically, JNK, and ERK were also reduced with FtL pre-treatment. Taken together, our findings elucidate a crucial immunomodulatory function for circulating ferritin that challenges the traditional view of this protein as a mere marker of body iron stores. Accordingly, these findings will stimulate investigations to the adaptive nature of this protein in diverse clinical settings.
Collapse
Affiliation(s)
- Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Laurence M. Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kayla R. McCullough
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Travis D. Hull
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephanie K. Esman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ravindra Boddu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | | - Wenguang Feng
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Paolo Arosio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jozsef Balla
- Department of Nephrology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Development and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
27
|
Lever JM, Hull TD, Boddu R, Pepin ME, Black LM, Adedoyin OO, Yang Z, Traylor AM, Jiang Y, Li Z, Peabody JE, Eckenrode HE, Crossman DK, Crowley MR, Bolisetty S, Zimmerman KA, Wende AR, Mrug M, Yoder BK, Agarwal A, George JF. Resident macrophages reprogram toward a developmental state after acute kidney injury. JCI Insight 2019; 4:e125503. [PMID: 30674729 PMCID: PMC6413788 DOI: 10.1172/jci.insight.125503] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Acute kidney injury (AKI) is a devastating clinical condition affecting at least two-thirds of critically ill patients, and, among these patients, it is associated with a greater than 60% risk of mortality. Kidney mononuclear phagocytes (MPs) are implicated in pathogenesis and healing in mouse models of AKI and, thus, have been the subject of investigation as potential targets for clinical intervention. We have determined that, after injury, F4/80hi-expressing kidney-resident macrophages (KRMs) are a distinct cellular subpopulation that does not differentiate from nonresident infiltrating MPs. However, if KRMs are depleted using polyinosinic/polycytidylic acid (poly I:C), they can be reconstituted from bone marrow-derived precursors. Further, KRMs lack major histocompatibility complex class II (MHCII) expression before P7 but upregulate it over the next 14 days. This MHCII- KRM phenotype reappears after injury. RNA sequencing shows that injury causes transcriptional reprogramming of KRMs such that they more closely resemble that found at P7. KRMs after injury are also enriched in Wingless-type MMTV integration site family (Wnt) signaling, indicating that a pathway vital for mouse and human kidney development is active. These data indicate that mechanisms involved in kidney development may be functioning after injury in KRMs.
Collapse
Affiliation(s)
- Jeremie M. Lever
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Travis D. Hull
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ravindra Boddu
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Laurence M. Black
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Oreoluwa O. Adedoyin
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zhengqin Yang
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amie M. Traylor
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yanlin Jiang
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zhang Li
- Department of Cellular, Developmental and Integrative Biology, and
| | | | - Han E. Eckenrode
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael R. Crowley
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Subhashini Bolisetty
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | - Michal Mrug
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Veterans Affairs, Birmingham, Alabama, USA
| | - Bradley K. Yoder
- Department of Cellular, Developmental and Integrative Biology, and
| | - Anupam Agarwal
- Department of Medicine and
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Veterans Affairs, Birmingham, Alabama, USA
| | - James F. George
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
28
|
Chinta KC, Rahman MA, Saini V, Glasgow JN, Reddy VP, Lever JM, Nhamoyebonde S, Leslie A, Wells RM, Traylor A, Madansein R, Siegal GP, Antony VB, Deshane J, Wells G, Nargan K, George JF, Ramdial PK, Agarwal A, Steyn AJC. Microanatomic Distribution of Myeloid Heme Oxygenase-1 Protects against Free Radical-Mediated Immunopathology in Human Tuberculosis. Cell Rep 2018; 25:1938-1952.e5. [PMID: 30428359 PMCID: PMC6250977 DOI: 10.1016/j.celrep.2018.10.073] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 07/18/2018] [Accepted: 10/19/2018] [Indexed: 11/26/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that controls inflammatory responses and redox homeostasis; however, its role during pulmonary tuberculosis (TB) remains unclear. Using freshly resected human TB lung tissue, we examined the role of HO-1 within the cellular and pathological spectrum of TB. Flow cytometry and histopathological analysis of human TB lung tissues showed that HO-1 is expressed primarily in myeloid cells and that HO-1 levels in these cells were directly proportional to cytoprotection. HO-1 mitigates TB pathophysiology by diminishing myeloid cell-mediated oxidative damage caused by reactive oxygen and/or nitrogen intermediates, which control granulocytic karyorrhexis to generate a zonal HO-1 response. Using whole-body or myeloid-specific HO-1-deficient mice, we demonstrate that HO-1 is required to control myeloid cell infiltration and inflammation to protect against TB progression. Overall, this study reveals that zonation of HO-1 in myeloid cells modulates free-radical-mediated stress, which regulates human TB immunopathology.
Collapse
Affiliation(s)
- Krishna C Chinta
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Vikram Saini
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Joel N Glasgow
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Vineel P Reddy
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremie M Lever
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | - Ryan M Wells
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amie Traylor
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Gene P Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Veena B Antony
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jessy Deshane
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gordon Wells
- Africa Health Research Institute, Durban 4001, South Africa
| | | | - James F George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pratistadevi K Ramdial
- Department of Anatomical Pathology, NHLS, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban 4091, South Africa
| | - Anupam Agarwal
- Nephrology Research and Training Center, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Veterans Affairs, Birmingham, AL 35294, USA
| | - Adrie J C Steyn
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Africa Health Research Institute, Durban 4001, South Africa; UAB Center for AIDS Research, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
29
|
Zhang M, Nakamura K, Kageyama S, Lawal AO, Gong KW, Bhetraratana M, Fujii T, Sulaiman D, Hirao H, Bolisetty S, Kupiec-Weglinski JW, Araujo JA. Myeloid HO-1 modulates macrophage polarization and protects against ischemia-reperfusion injury. JCI Insight 2018; 3:120596. [PMID: 30282830 DOI: 10.1172/jci.insight.120596] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/21/2018] [Indexed: 01/23/2023] Open
Abstract
Macrophages polarize into heterogeneous proinflammatory M1 and antiinflammatory M2 subtypes. Heme oxygenase 1 (HO-1) protects against inflammatory processes such as ischemia-reperfusion injury (IRI), organ transplantation, and atherosclerosis. To test our hypothesis that HO-1 regulates macrophage polarization and protects against IRI, we generated myeloid-specific HO-1-knockout (mHO-1-KO) and -transgenic (mHO-1-Tg) mice, with deletion or overexpression of HO-1, in various macrophage populations. Bone marrow-derived macrophages (BMDMs) from mHO-1-KO mice, treated with M1-inducing LPS or M2-inducing IL-4, exhibited increased mRNA expression of M1 (CXCL10, IL-1β, MCP1) and decreased expression of M2 (Arg1 and CD163) markers as compared with controls, while BMDMs from mHO-1-Tg mice displayed the opposite. A similar pattern was observed in the hepatic M1/M2 expression profile in a mouse model of liver IRI. mHO-1-KO mice displayed increased hepatocellular damage, serum AST/ALT levels, Suzuki's histological score of liver IRI, and neutrophil and macrophage infiltration, while mHO-1-Tg mice exhibited the opposite. In human liver transplant biopsies, subjects with higher HO-1 levels showed lower expression of M1 markers together with decreased hepatocellular damage and improved outcomes. In conclusion, myeloid HO-1 expression modulates macrophage polarization, and protects against liver IRI, at least in part by favoring an M2 phenotype.
Collapse
Affiliation(s)
- Min Zhang
- Department of Medicine, Division of Cardiology, and
| | - Kojiro Nakamura
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Shoichi Kageyama
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Ke Wei Gong
- Department of Medicine, Division of Cardiology, and
| | | | - Takehiro Fujii
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Hirofumi Hirao
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Jerzy W Kupiec-Weglinski
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jesus A Araujo
- Department of Medicine, Division of Cardiology, and.,Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles, California, USA
| |
Collapse
|
30
|
Dual effect of hemin on renal ischemia-reperfusion injury. Biochem Biophys Res Commun 2018; 503:2820-2825. [PMID: 30100067 DOI: 10.1016/j.bbrc.2018.08.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 01/07/2023]
Abstract
Acute kidney injury (AKI) is a major public health concern, which is contributing to serious hospital complications, chronic kidney disease (CKD) and even death. Renal ischemia-reperfusion injury (IRI) remains a leading cause of AKI. The stress-responsive enzyme, heme oxygenase-1 (HO-1) mediates protection against renal IRI and may be preventively induced using hemin prior to renal insult. This HO-1 induction pathway called hemin preconditioning is largely known to be effective. Therefore, HO-1 might be an interesting therapeutic target in case of predictable AKI (e.g. partial nephrectomy or renal transplantation). However, the use of hemin to mitigate established AKI remains poorly characterized. Mice underwent bilateral renal IRI for 26 min or sham surgery. After surgical procedure, animals were injected either with hemin (5 mg/kg) or vehicle. Twenty-four hours later, mice were sacrificed. Despite strong HO-1 induction, hemin-treated mice exhibited significant renal damage and oxidative stress as compared to vehicle-treated mice. Interestingly, higher dose of hemin is associated with more severe IRI-induced AKI in a dose-dependent relation. To determine whether hemin preconditioning remains efficient to dampen postoperative hemin-amplified IRI-induced AKI, we pretreated mice either with hemin (5 mg/kg) or vehicle 24 h prior to surgical procedure. Then, all mice (hemin- and vehicle-pretreated) received postoperative injection of hemin (5 mg/kg) to amplify IRI-induced AKI. In comparison to vehicle, prior administration of hemin to renal IRI mitigated hemin-amplified IRI-induced AKI as attested by fewer renal damage, inflammation and oxidative stress. In conclusion, hemin may have a dual effect on renal IRI, protective or deleterious, depending on the timing of its administration.
Collapse
|
31
|
Black LM, Lever JM, Traylor AM, Chen B, Yang Z, Esman SK, Jiang Y, Cutter GR, Boddu R, George JF, Agarwal A. Divergent effects of AKI to CKD models on inflammation and fibrosis. Am J Physiol Renal Physiol 2018; 315:F1107-F1118. [PMID: 29897282 DOI: 10.1152/ajprenal.00179.2018] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic kidney disease (CKD) is a condition with significant morbidity and mortality that affects 15% of adults in the United States. One cause of CKD is acute kidney injury (AKI), which commonly occurs secondary to sepsis, ischemic events, and drug-induced nephrotoxicity. Unilateral ischemia-reperfusion injury (UIRI) without contralateral nephrectomy (CLN) and repeated low-dose cisplatin (RLDC) models of AKI to CKD demonstrate responses characteristic of the transition; however, previous studies have not effectively compared the pathogenesis. We demonstrate both models instigate renal dysfunction, inflammatory cytokine responses, and fibrosis. However, the models exhibit differences in urinary excretory function, inflammatory cell infiltration, and degree of fibrotic response. UIRI without CLN demonstrated worsening perfusion and function, measured with 99mTc-mercaptoacetyltriglycine-3 imaging, and physiologic compensation in the contralateral kidney. Furthermore, UIRI without CLN elicited a robust inflammatory response that was characterized by a prolonged polymorphonuclear cell and natural killer cell infiltrate and an early expansion of kidney resident macrophages, followed by T-cell infiltration. Symmetrical diminished function occurred in RLDC kidneys and progressively worsened until day 17 of the study. Surprisingly, RLDC mice demonstrated a decrease in inflammatory cell numbers relative to controls. However, RLDC kidneys expressed increased levels of kidney injury molecule-1 (KIM-1), high mobility group box-1 ( HMGB1), and colony stimulating factor-1 ( CSF-1), which likely recruits inflammatory cells in response to injury. These data emphasize how the divergent etiologies of AKI to CKD models affect the kidney microenvironment and outcomes. This study provides support for subtyping AKI by etiology in human studies, aiding in the elucidation of injury-specific pathophysiologic mechanisms of the AKI to CKD transition.
Collapse
Affiliation(s)
- L M Black
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - J M Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - A M Traylor
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - B Chen
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Z Yang
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - S K Esman
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Y Jiang
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - G R Cutter
- Department of Biostatistics, University of Alabama at Birmingham , Birmingham, Alabama
| | - R Boddu
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - J F George
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Surgery, University of Alabama at Birmingham , Birmingham, Alabama
| | - A Agarwal
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Veterans Affairs , Birmingham, Alabama
| |
Collapse
|
32
|
Parabiosis reveals leukocyte dynamics in the kidney. J Transl Med 2018; 98:391-402. [PMID: 29251733 PMCID: PMC5839939 DOI: 10.1038/labinvest.2017.130] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/10/2017] [Accepted: 09/21/2017] [Indexed: 12/17/2022] Open
Abstract
The immune cellular compartment of the kidney is involved in organ development and homeostasis, as well as in many pathological conditions. Little is known about the mechanisms that drive intrarenal immune responses in the presence of renal tubular and interstitial cell death. However, it is known that tissue-resident leukocytes have the potential to have distinct roles compared with circulating cells. We used a parabiosis model in C57BL/6 CD45 congenic and green fluorescent protein transgenic mice to better understand the dynamics of immune cells in the kidney. We found F4/80Hi intrarenal macrophages exhibit minimal exchange with the peripheral circulation in two models of parabiosis, whether mice were attached for 4 or 16 weeks. Other intrarenal inflammatory cells demonstrate near total exchange with the circulating immune cell pool in healthy kidneys, indicating that innate and adaptive immune cells extensively traffic through the kidney interstitium during normal physiology. Neutrophils, dendritic cells, F4/80Low macrophages, T cells, B cells, and NK cells are renewed from the circulating immune cell pool. However, a fraction of double-negative T (CD4- CD8-) and NKT cells are long-lived or tissue resident. This study provides direct evidence of leukocyte sub-populations that are resident in the renal tissue, cells which demonstrate minimal to no exchange with the peripheral blood. In addition, the data demonstrate continual exchange of other sub-populations through uninflamed tissue.
Collapse
|
33
|
Abstract
Acute kidney injury (AKI) is a growing global health concern, yet no treatment is currently available to prevent it or to promote kidney repair after injury. Animal models demonstrate that the macrophage is a major contributor to the inflammatory response to AKI. Emerging data from human biopsies also corroborate the presence of macrophages in AKI and their persistence in progressive chronic kidney disease. Macrophages are phagocytic innate immune cells that are important mediators of tissue homeostasis and host defense. In response to tissue injury, macrophages become activated based on specific signals from the damaged microenvironment. The activation and functional state of the macrophage depends on the stage of tissue injury and repair, reflecting a dynamic and diverse spectrum of macrophage phenotypes. In this review, we highlight our current understanding of the mechanisms by which macrophages contribute to injury and repair after AKI.
Collapse
Affiliation(s)
- Sarah C Huen
- Section of Nephrology, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520;
| | - Lloyd G Cantley
- Section of Nephrology, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520;
| |
Collapse
|
34
|
Song CJ, Zimmerman KA, Henke SJ, Yoder BK. Inflammation and Fibrosis in Polycystic Kidney Disease. Results Probl Cell Differ 2017; 60:323-344. [PMID: 28409351 PMCID: PMC7875307 DOI: 10.1007/978-3-319-51436-9_12] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Polycystic kidney disease (PKD) is a commonly inherited disorder characterized by cyst formation and fibrosis (Wilson, N Engl J Med 350:151-164, 2004) and is caused by mutations in cilia or cilia-related proteins, such as polycystin 1 or 2 (Oh and Katsanis, Development 139:443-448, 2012; Kotsis et al., Nephrol Dial Transplant 28:518-526, 2013). A major pathological feature of PKD is the development of interstitial inflammation and fibrosis with an associated accumulation of inflammatory cells (Grantham, N Engl J Med 359:1477-1485, 2008; Zeier et al., Kidney Int 42:1259-1265, 1992; Ibrahim, Sci World J 7:1757-1767, 2007). It is unclear whether inflammation is a driving force for cyst formation or a consequence of the pathology (Ta et al., Nephrology 18:317-330, 2013) as in some murine models cysts are present prior to the increase in inflammatory cells (Phillips et al., Kidney Blood Press Res 30:129-144, 2007; Takahashi et al., J Am Soc Nephrol JASN 1:980-989, 1991), while in other models the increase in inflammatory cells is present prior to or coincident with cyst initiation (Cowley et al., Kidney Int 43:522-534, 1993, Kidney Int 60:2087-2096, 2001). Additional support for inflammation as an important contributor to cystic kidney disease is the increased expression of many pro-inflammatory cytokines in murine models and human patients with cystic kidney disease (Karihaloo et al., J Am Soc Nephrol JASN 22:1809-1814, 2011; Swenson-Fields et al., Kidney Int, 2013; Li et al., Nat Med 14:863-868, 2008a). Based on these data, an emerging model in the field is that disruption of primary cilia on tubule epithelial cells leads to abnormal cytokine cross talk between the epithelium and the inflammatory cells contributing to cyst growth and fibrosis (Ta et al., Nephrology 18:317-330, 2013). These cytokines are produced by interstitial fibroblasts, inflammatory cells, and tubule epithelial cells and activate multiple pathways including the JAK-STAT and NF-κB signaling (Qin et al., J Am Soc Nephrol JASN 23:1309-1318, 2012; Park et al., Am J Nephrol 32:169-178, 2010; Bhunia et al., Cell 109:157-168, 2002). Indeed, inflammatory cells are responsible for producing several of the pro-fibrotic growth factors observed in PKD patients with fibrosis (Nakamura et al., Am J Nephrol 20:32-36, 2000; Wilson et al., J Cell Physiol 150:360-369, 1992; Song et al., Hum Mol Genet 18:2328-2343, 2009; Schieren et al., Nephrol Dial Transplant 21:1816-1824, 2006). These growth factors trigger epithelial cell proliferation and myofibroblast activation that stimulate the production of extracellular matrix (ECM) genes including collagen types 1 and 3 and fibronectin, leading to reduced glomerular function with approximately 50% of ADPKD patients progressing to end-stage renal disease (ESRD). Therefore, treatments designed to reduce inflammation and slow the rate of fibrosis are becoming important targets that hold promise to improve patient life span and quality of life. In fact, recent studies in several PKD mouse models indicate that depletion of macrophages reduces cyst severity. In this chapter, we review the potential mechanisms of interstitial inflammation in PKD with a focus on ADPKD and discuss the role of interstitial inflammation in progression to fibrosis and ESRD.
Collapse
Affiliation(s)
- Cheng Jack Song
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kurt A Zimmerman
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Scott J Henke
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bradley K Yoder
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
35
|
Boddu R, Fan C, Rangarajan S, Sunil B, Bolisetty S, Curtis LM. Unique sex- and age-dependent effects in protective pathways in acute kidney injury. Am J Physiol Renal Physiol 2017; 313:F740-F755. [PMID: 28679590 PMCID: PMC5625098 DOI: 10.1152/ajprenal.00049.2017] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/06/2017] [Accepted: 06/28/2017] [Indexed: 12/25/2022] Open
Abstract
Sex and age influence susceptibility to acute kidney injury (AKI), with young females exhibiting lowest incidence. In these studies, we investigated mechanisms which may underlie the sex/age-based dissimilarities. Cisplatin (Cp)-induced AKI resulted in morphological evidence of injury in all groups. A minimal rise in plasma creatinine (PCr) was seen in Young Females, whereas in Aged Females, PCr rose precipitously. Relative to Young Males, Aged Males showed significantly, but temporally, comparably elevated PCr. Notably, Aged Females showed significantly greater mortality, whereas Young Females exhibited none. Tissue KIM-1 and plasma NGAL were significantly lower in Young Females than all others. IGFBP7 levels were modestly increased in both Young groups. IGFBP7 levels in Aged Females were significantly elevated at baseline relative to Aged Males, and increased linearly through day 3, when these levels were comparable in both Aged groups. Plasma cytokine levels similarly showed a pattern of protective effects preferentially in Young Females. Expression of the drug transporter MATE2 did not explain the sex/age distinctions. Heme oxygenase-1 (HO-1) levels (~28-kDa species) showed elevation at day 1 in all groups with highest levels seen in Young Males. Exclusively in Young Females, these levels returned to baseline on day 3, suggestive of a more efficient recovery. In aggregate, we demonstrate, for the first time, a distinctive pattern of response to AKI in Young Females relative to males which appears to be significantly altered in aging. These distinctions may offer novel targets to exploit therapeutically in both females and males in the treatment of AKI.
Collapse
Affiliation(s)
- Ravindra Boddu
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Chunlan Fan
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Sunil Rangarajan
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Bhuvana Sunil
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Subhashini Bolisetty
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Lisa M Curtis
- Division of Nephrology, Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and .,Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
36
|
Adedoyin O, Boddu R, Traylor A, Lever JM, Bolisetty S, George JF, Agarwal A. Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol Renal Physiol 2017; 314:F702-F714. [PMID: 28515173 DOI: 10.1152/ajprenal.00044.2017] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI because of its antiapoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of antioxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared with vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Cotreatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-l-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important antiferroptotic role of HO-1 in renal epithelial cells.
Collapse
Affiliation(s)
- Oreoluwa Adedoyin
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Ravindra Boddu
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Amie Traylor
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Jeremie M Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Subhashini Bolisetty
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - James F George
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Surgery, University of Alabama at Birmingham , Birmingham, Alabama
| | - Anupam Agarwal
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Birmingham VA Medical Center , Birmingham, Alabama
| |
Collapse
|
37
|
Specific expression of heme oxygenase-1 by myeloid cells modulates renal ischemia-reperfusion injury. Sci Rep 2017; 7:197. [PMID: 28298633 PMCID: PMC5428056 DOI: 10.1038/s41598-017-00220-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 02/14/2017] [Indexed: 12/21/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a major risk factor for delayed graft function in renal transplantation. Compelling evidence exists that the stress-responsive enzyme, heme oxygenase-1 (HO-1) mediates protection against IRI. However, the role of myeloid HO-1 during IRI remains poorly characterized. Mice with myeloid-restricted deletion of HO-1 (HO-1M-KO), littermate (LT), and wild-type (WT) mice were subjected to renal IRI or sham procedures and sacrificed after 24 hours or 7 days. In comparison to LT, HO-1M-KO exhibited significant renal histological damage, pro-inflammatory responses and oxidative stress 24 hours after reperfusion. HO-1M-KO mice also displayed impaired tubular repair and increased renal fibrosis 7 days after IRI. In WT mice, HO-1 induction with hemin specifically upregulated HO-1 within the CD11b+ F4/80lo subset of the renal myeloid cells. Prior administration of hemin to renal IRI was associated with significant increase of the renal HO-1+ CD11b+ F4/80lo myeloid cells in comparison to control mice. In contrast, this hemin-mediated protection was abolished in HO-1M-KO mice. In conclusion, myeloid HO-1 appears as a critical protective pathway against renal IRI and could be an interesting therapeutic target in renal transplantation.
Collapse
|
38
|
Winfree S, Khan S, Micanovic R, Eadon MT, Kelly KJ, Sutton TA, Phillips CL, Dunn KW, El-Achkar TM. Quantitative Three-Dimensional Tissue Cytometry to Study Kidney Tissue and Resident Immune Cells. J Am Soc Nephrol 2017; 28:2108-2118. [PMID: 28154201 DOI: 10.1681/asn.2016091027] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022] Open
Abstract
Analysis of the immune system in the kidney relies predominantly on flow cytometry. Although powerful, the process of tissue homogenization necessary for flow cytometry analysis introduces bias and results in the loss of morphologic landmarks needed to determine the spatial distribution of immune cells. An ideal approach would support three-dimensional (3D) tissue cytometry: an automated quantitation of immune cells and associated spatial parameters in 3D image volumes collected from intact kidney tissue. However, widespread application of this approach is limited by the lack of accessible software tools for digital analysis of large 3D microscopy data. Here, we describe Volumetric Tissue Exploration and Analysis (VTEA) image analysis software designed for efficient exploration and quantitative analysis of large, complex 3D microscopy datasets. In analyses of images collected from fixed kidney tissue, VTEA replicated the results of flow cytometry while providing detailed analysis of the spatial distribution of immune cells in different regions of the kidney and in relation to specific renal structures. Unbiased exploration with VTEA enabled us to discover a population of tubular epithelial cells that expresses CD11C, a marker typically expressed on dendritic cells. Finally, we show the use of VTEA for large-scale quantitation of immune cells in entire human kidney biopsies. In summary, we show that VTEA is a simple and effective tool that supports unique digital interrogation and analysis of kidney tissue from animal models or biobanked human kidney biopsies. We have made VTEA freely available to interested investigators via electronic download.
Collapse
Affiliation(s)
- Seth Winfree
- Division of Nephrology, Department of Medicine and
| | - Shehnaz Khan
- Division of Nephrology, Department of Medicine and
| | | | | | | | | | - Carrie L Phillips
- Department of Pathology, Indiana University School of Medicine, Indianapolis, Indiana; and
| | | | - Tarek M El-Achkar
- Division of Nephrology, Department of Medicine and .,Department of Medicine, Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| |
Collapse
|
39
|
Bolisetty S, Zarjou A, Agarwal A. Heme Oxygenase 1 as a Therapeutic Target in Acute Kidney Injury. Am J Kidney Dis 2017; 69:531-545. [PMID: 28139396 DOI: 10.1053/j.ajkd.2016.10.037] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/22/2016] [Indexed: 01/06/2023]
Abstract
A common clinical condition, acute kidney injury (AKI) significantly influences morbidity and mortality, particularly in critically ill patients. The pathophysiology of AKI is complex and involves multiple pathways, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Recent evidence suggests that a single insult to the kidney significantly enhances the propensity to develop chronic kidney disease. Therefore, the generation of effective therapies against AKI is timely. In this context, the cytoprotective effects of heme oxygenase 1 (HO-1) in animal models of AKI are well documented. HO-1 modulates oxidative stress, autophagy, and inflammation and regulates the progression of cell cycle via direct and indirect mechanisms. These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). This review highlights recent advances in the molecular mechanisms of HO-1-mediated cytoprotection and discusses the translational potential of HO-1 induction in AKI.
Collapse
Affiliation(s)
- Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL; Birmingham Veterans Administration Medical Center, Birmingham, AL.
| |
Collapse
|
40
|
George JF, Lever JM, Agarwal A. Mononuclear phagocyte subpopulations in the mouse kidney. Am J Physiol Renal Physiol 2017; 312:F640-F646. [PMID: 28100500 DOI: 10.1152/ajprenal.00369.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 12/13/2022] Open
Abstract
Mononuclear phagocytes are the most common cells in the kidney associated with immunity and inflammation. Although the presence of these cells in the kidney has been known for decades, the study of mononuclear phagocytes in the context of kidney function and dysfunction is still at an early stage. The purpose of this review is to summarize the present knowledge regarding classification of these cells in the mouse kidney and to identify relevant questions that would further advance the field and potentially lead to new opportunities for treatment of acute kidney injury and other kidney diseases.
Collapse
Affiliation(s)
- James F George
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Nephrology Research and Training Center; University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Jeremie M Lever
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Nephrology Research and Training Center; University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; .,Department of Nephrology Research and Training Center; University of Alabama at Birmingham, Birmingham, Alabama; and.,Department of Veterans Affairs, Birmingham, Alabama
| |
Collapse
|
41
|
Abstract
Iron is an essential metal involved in several major cellular processes required to maintain life. Because of iron's ability to cause oxidative damage, its transport, metabolism, and storage is strictly controlled in the body, especially in the small intestine, liver, and kidney. Iron plays a major role in acute kidney injury and has been a target for therapeutic intervention. However, the therapies that have been effective in animal models of acute kidney injury have not been successful in human beings. Targeting iron trafficking via ferritin, ferroportin, or hepcidin may offer new insights. This review focuses on the biology of iron, particularly in the kidney, and its implications in acute kidney injury.
Collapse
Affiliation(s)
- Vyvyca J Walker
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Birmingham Veterans Administration Medical Center, Birmingham, AL.
| |
Collapse
|
42
|
Abstract
SIGNIFICANCE Acute kidney injury (AKI) and chronic kidney disease (CKD) represent a considerable burden in healthcare. The heme oxygenase (HO) system plays an important role in regulating oxidative stress and is protective in a variety of human and animal models of kidney disease. Preclinical studies of the HO system have led to the development of several clinical trials targeting the enzyme or its products. RECENT ADVANCES Connection of HO, ferritin, and other proteins involved in iron regulation has provided important insight into mechanisms of damage in AKI. Also, HO-1 expression is important in the pathogenesis of hypertension, diabetic kidney disease, and progression to end-stage renal disease. CRITICAL ISSUES Despite intriguing discoveries, no drugs targeting the HO system have been translated to the clinic. Meanwhile, treatments for AKI and CKD are urgently needed. Many factors have likely contributed to challenges in clinical translation, including variation in animal models, difficulties in obtaining human tissue, and complexity of the disease processes being studied. FUTURE DIRECTIONS The HO system represents a promising avenue of investigation that may lead to targeted therapeutics. Tissue-specific gene modulation, widening the scope of animal studies, and continued clinical research will provide valuable insight into the role HO plays in kidney homeostasis and disease. Antioxid. Redox Signal. 25, 165-183.
Collapse
Affiliation(s)
- Jeremie M Lever
- 1 Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham , Birmingham, Alabama
| | - Ravindra Boddu
- 1 Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham , Birmingham, Alabama
| | - James F George
- 2 Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham , Birmingham, Alabama
| | - Anupam Agarwal
- 1 Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham , Birmingham, Alabama.,3 Birmingham Veterans Administration Medical Center , Birmingham, Alabama
| |
Collapse
|
43
|
Leaf DE, Body SC, Muehlschlegel JD, McMahon GM, Lichtner P, Collard CD, Shernan SK, Fox AA, Waikar SS. Length Polymorphisms in Heme Oxygenase-1 and AKI after Cardiac Surgery. J Am Soc Nephrol 2016; 27:3291-3297. [PMID: 27257045 DOI: 10.1681/asn.2016010038] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/28/2016] [Indexed: 11/03/2022] Open
Abstract
Heme oxygenase-1 (HO-1) catalyzes the degradation of heme, which may be involved in the pathogenesis of AKI. Length polymorphisms in the number of GT dinucleotide repeats in the HO-1 gene (HMOX1) promoter inversely associate with HMOX1 mRNA expression. We analyzed the association between allelic frequencies of GT repeats in the HMOX1 gene promoter and postoperative AKI in 2377 white patients who underwent cardiac surgery with cardiopulmonary bypass. We categorized patients as having the short allele (S; <27 GT repeats) or long allele (L; ≥27 GT repeats), and defined AKI as an increase in serum creatinine ≥0.3 mg/dl within 48 hours or ≥50% within 5 days, or the need for RRT. Compared with patients with the SS genotype, patients with the LL genotype had 1.58-fold (95% confidence interval, 1.06 to 2.34; P=0.02) higher odds of AKI. After adjusting for baseline and operative characteristics, the odds ratio for AKI per L allele was 1.26 (95% confidence interval, 1.05 to 1.50; P=0.01). In conclusion, longer GT repeats in the HMOX1 gene promoter associate with increased risk of AKI after cardiac surgery, consistent with heme toxicity as a pathogenic feature of cardiac surgery-associated AKI, and with HO-1 as a potential therapeutic target.
Collapse
Affiliation(s)
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Peter Lichtner
- Genome Analysis Center, Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Charles D Collard
- Department of Anesthesiology, Baylor St. Luke's Medical Center and the Texas Heart Institute, Houston, Texas; and
| | - Stanton K Shernan
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Amanda A Fox
- Department of Anesthesiology and Pain Management, and McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|
44
|
Yang Y, Song M, Liu Y, Liu H, Sun L, Peng Y, Liu F, Venkatachalam MA, Dong Z. Renoprotective approaches and strategies in acute kidney injury. Pharmacol Ther 2016; 163:58-73. [PMID: 27108948 DOI: 10.1016/j.pharmthera.2016.03.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/18/2016] [Indexed: 12/17/2022]
Abstract
Acute kidney injury (AKI) is a major renal disease associated with high mortality rate and increasing prevalence. Decades of research have suggested numerous chemical and biological agents with beneficial effects in AKI. In addition, cell therapy and molecular targeting have been explored for reducing kidney tissue damage and promoting kidney repair or recovery from AKI. Mechanistically, these approaches may mitigate oxidative stress, inflammation, cell death, and mitochondrial and other organellar damage, or activate cytoprotective mechanisms such as autophagy and pro-survival factors. However, none of these findings has been successfully translated into clinical treatment of AKI. In this review, we analyze these findings and propose experimental strategies for the identification of renoprotective agents or methods with clinical potential. Moreover, we propose the consideration of combination therapy by targeting multiple targets in AKI.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meifang Song
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youming Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | | | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
45
|
Inhibition of Ectodermal-Neural Cortex 1 Protects Neural Cells from Apoptosis Induced by Hypoxia and Hypoglycemia. J Mol Neurosci 2016; 59:126-34. [DOI: 10.1007/s12031-016-0742-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
|
46
|
Bolisetty S, Traylor A, Joseph R, Zarjou A, Agarwal A. Proximal tubule-targeted heme oxygenase-1 in cisplatin-induced acute kidney injury. Am J Physiol Renal Physiol 2016; 310:F385-94. [PMID: 26672618 PMCID: PMC4868370 DOI: 10.1152/ajprenal.00335.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that catalyzes the breakdown of heme to biliverdin, carbon monoxide, and iron. The beneficial effects of HO-1 expression are not merely due to degradation of the pro-oxidant heme but are also credited to the by-products that have potent, protective effects, including antioxidant, anti-inflammatory, and prosurvival properties. This is well reflected in the preclinical animal models of injury in both renal and nonrenal settings. However, excessive accumulation of the by-products can be deleterious and lead to mitochondrial toxicity and oxidative stress. Therefore, use of the HO system in alleviating injury merits a targeted approach. Based on the higher susceptibility of the proximal tubule segment of the nephron to injury, we generated transgenic mice using cre-lox technology to enable manipulation of HO-1 (deletion or overexpression) in a cell-specific manner. We demonstrate the validity and feasibility of these mice by breeding them with proximal tubule-specific Cre transgenic mice. Similar to previous reports using chemical modulators and global transgenic mice, we demonstrate that whereas deletion of HO-1, specifically in the proximal tubules, aggravates structural and functional damage during cisplatin nephrotoxicity, selective overexpression of HO-1 in proximal tubules is protective. At the cellular level, cleaved caspase-3 expression, a marker of apoptosis, and p38 signaling were modulated by HO-1. Use of these transgenic mice will aid in the evaluation of the effects of cell-specific HO-1 expression in response to injury and assist in the generation of targeted approaches that will enhance recovery with reduced, unwarranted adverse effects.
Collapse
Affiliation(s)
- Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Amie Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Reny Joseph
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; and Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
47
|
Hull TD, Boddu R, Guo L, Tisher CC, Traylor AM, Patel B, Joseph R, Prabhu SD, Suliman HB, Piantadosi CA, Agarwal A, George JF. Heme oxygenase-1 regulates mitochondrial quality control in the heart. JCI Insight 2016; 1:e85817. [PMID: 27110594 DOI: 10.1172/jci.insight.85817] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The cardioprotective inducible enzyme heme oxygenase-1 (HO-1) degrades prooxidant heme into equimolar quantities of carbon monoxide, biliverdin, and iron. We hypothesized that HO-1 mediates cardiac protection, at least in part, by regulating mitochondrial quality control. We treated WT and HO-1 transgenic mice with the known mitochondrial toxin, doxorubicin (DOX). Relative to WT mice, mice globally overexpressing human HO-1 were protected from DOX-induced dilated cardiomyopathy, cardiac cytoarchitectural derangement, and infiltration of CD11b+ mononuclear phagocytes. Cardiac-specific overexpression of HO-1 ameliorated DOX-mediated dilation of the sarcoplasmic reticulum as well as mitochondrial disorganization in the form of mitochondrial fragmentation and increased numbers of damaged mitochondria in autophagic vacuoles. HO-1 overexpression promotes mitochondrial biogenesis by upregulating protein expression of NRF1, PGC1α, and TFAM, which was inhibited in WT animals treated with DOX. Concomitantly, HO-1 overexpression inhibited the upregulation of the mitochondrial fission mediator Fis1 and resulted in increased expression of the fusion mediators, Mfn1 and Mfn2. It also prevented dynamic changes in the levels of key mediators of the mitophagy pathway, PINK1 and parkin. Therefore, these findings suggest that HO-1 has a novel role in protecting the heart from oxidative injury by regulating mitochondrial quality control.
Collapse
Affiliation(s)
- Travis D Hull
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ravindra Boddu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lingling Guo
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cornelia C Tisher
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bindiya Patel
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Reny Joseph
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sumanth D Prabhu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Veterans Affairs, Birmingham, Alabama, USA
| | - Hagir B Suliman
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Claude A Piantadosi
- Department of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Veterans Affairs, Birmingham, Alabama, USA
| | - James F George
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
48
|
Rao S, Walters KB, Wilson L, Chen B, Bolisetty S, Graves D, Barnes S, Agarwal A, Kabarowski JH. Early lipid changes in acute kidney injury using SWATH lipidomics coupled with MALDI tissue imaging. Am J Physiol Renal Physiol 2016; 310:F1136-47. [PMID: 26911846 DOI: 10.1152/ajprenal.00100.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 02/23/2016] [Indexed: 11/22/2022] Open
Abstract
Acute kidney injury (AKI) is one of the leading causes of in-hospital morbidity and mortality, particularly in critically ill patients. Although our understanding of AKI at the molecular level remains limited due to its complex pathophysiology, recent advances in both quantitative and spatial mass spectrometric approaches offer new opportunities to assess the significance of renal metabolomic changes in AKI models. In this study, we evaluated lipid changes in early ischemia-reperfusion (IR)-related AKI in mice by using sequential window acquisition of all theoretical spectra (SWATH)-mass spectrometry (MS) lipidomics. We found a significant increase in two abundant ether-linked phospholipids following IR at 6 h postinjury, a plasmanyl choline, phosphatidylcholine (PC) O-38:1 (O-18:0, 20:1), and a plasmalogen, phosphatidylethanolamine (PE) O-42:3 (O-20:1, 22:2). Both of these lipids correlated with the severity of AKI as measured by plasma creatinine. In addition to many more renal lipid changes associated with more severe AKI, PC O-38:1 elevations were maintained at 24 h post-IR, while renal PE O-42:3 levels decreased, as were all ether PEs detected by SWATH-MS at this later time point. To further assess the significance of this early increase in PC O-38:1, we used matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS) to determine that it occurred in proximal tubules, a region of the kidney that is most prone to IR injury and also rich in the rate-limiting enzymes involved in ether-linked phospholipid biosynthesis. Use of SWATH-MS lipidomics in conjunction with MALDI-IMS for lipid localization will help in elucidating the role of lipids in the pathobiology of AKI.
Collapse
Affiliation(s)
- Sangeetha Rao
- Division of Pediatric Critical Care, Children's Hospital of Alabama, Birmingham, Alabama
| | - Kelly B Walters
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama; Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Landon Wilson
- Targeted Metabolomics and Proteomics Laboratory, Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Bo Chen
- Division of Nephrology and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Subhashini Bolisetty
- Division of Nephrology and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - David Graves
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephen Barnes
- Targeted Metabolomics and Proteomics Laboratory, Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Anupam Agarwal
- Division of Nephrology and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Janusz H Kabarowski
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama;
| |
Collapse
|
49
|
Hirsch S, El-Achkar T, Robbins L, Basta J, Heitmeier M, Nishinakamura R, Rauchman M. A mouse model of Townes-Brocks syndrome expressing a truncated mutant Sall1 protein is protected from acute kidney injury. Am J Physiol Renal Physiol 2015; 309:F852-63. [DOI: 10.1152/ajprenal.00222.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/21/2015] [Indexed: 11/22/2022] Open
Abstract
It has been postulated that developmental pathways are reutilized during repair and regeneration after injury, but functional analysis of many genes required for kidney formation has not been performed in the adult organ. Mutations in SALL1 cause Townes-Brocks syndrome (TBS) and nonsyndromic congenital anomalies of the kidney and urinary tract, both of which lead to childhood kidney failure. Sall1 is a transcriptional regulator that is expressed in renal progenitor cells and developing nephrons in the embryo. However, its role in the adult kidney has not been investigated. Using a mouse model of TBS ( Sall1 TBS), we investigated the role of Sall1 in response to acute kidney injury. Our studies revealed that Sall1 is expressed in terminally differentiated renal epithelia, including the S3 segment of the proximal tubule, in the mature kidney. Sall1 TBS mice exhibited significant protection from ischemia-reperfusion injury and aristolochic acid-induced nephrotoxicity. This protection from acute injury is seen despite the presence of slowly progressive chronic kidney disease in Sall1 TBS mice. Mice containing null alleles of Sall1 are not protected from acute kidney injury, indicating that expression of a truncated mutant protein from the Sall1 TBS allele, while causative of congenital anomalies, protects the adult kidney from injury. Our studies further revealed that basal levels of the preconditioning factor heme oxygenase-1 are elevated in Sall1 TBS kidneys, suggesting a mechanism for the relative resistance to injury in this model. Together, these studies establish a functional role for Sall1 in the response of the adult kidney to acute injury.
Collapse
Affiliation(s)
- Sara Hirsch
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri
- John Cochran Division, Veterans Affairs St. Louis Health Care System, St. Louis, Missouri
| | - Tarek El-Achkar
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Lynn Robbins
- Department of Internal Medicine (Nephrology), Saint Louis University, St. Louis, Missouri
- John Cochran Division, Veterans Affairs St. Louis Health Care System, St. Louis, Missouri
| | - Jeannine Basta
- Department of Internal Medicine (Nephrology), Saint Louis University, St. Louis, Missouri
- John Cochran Division, Veterans Affairs St. Louis Health Care System, St. Louis, Missouri
| | - Monique Heitmeier
- Department of Internal Medicine (Nephrology), Saint Louis University, St. Louis, Missouri
| | - Ryuichi Nishinakamura
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Michael Rauchman
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri
- Department of Internal Medicine (Nephrology), Saint Louis University, St. Louis, Missouri
- John Cochran Division, Veterans Affairs St. Louis Health Care System, St. Louis, Missouri
| |
Collapse
|
50
|
Affiliation(s)
- Gilbert R Kinsey
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|