1
|
Lee E, Ahn JH, Kang BC, Lee HS. Nrf2-Dependent Adaptation to Oxidative Stress Protects Against Progression of Diabetic Nephropathy. Antioxid Redox Signal 2025; 42:751-766. [PMID: 39531219 DOI: 10.1089/ars.2023.0431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Aims: Adaptation to oxidative stress is essential for maintaining protein and redox homeostasis in mammalian cells. Palmitic acid (PA) plays a central role in oxidative stress and immunoproteasome regulation in podocytes and diabetes, and eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have beneficial impact on diabetes. The role of Nrf2 in adaptation to oxidative stress and regulation of immunoproteasome by PA and EPA/DHA in podocytes and diabetic kidneys is not well defined. The present study describes the effect of PA- and EPA/DHA-induced oxidative stress in regulating Nrf2/immuoproteasome pathway in a model system relevant to diabetic nephropathy (DN). Results: Short PA exposure to podocytes promotes the upregulation of antioxidant proteins and immunoproteasome mediated by Nrf2, leading to acute transient oxidative stress adaptation. Both short- and long-term incubation of EPA or DHA in podocytes induced oxidative stress and activation of Nrf2, causing persistent oxidative stress adaptation. Long PA exposure to podocytes decreased the Nrf2 activity, and EPA/DHA attenuated these effects of PA. In db/db mice, feeding of EPA/DHA-rich fish oil increased oxidative stress in kidneys and induced renal cortical Nrf2 nuclear translocation and immunoproteasome overexpression, inhibiting the progression of DN. Innovation and Conclusion: We demonstrate an oxidative stress adaptation mechanism by PA and EPA/DHA regulated by Nrf2 in podocytes and kidneys of type 2 diabetes. This work provides an important insight into the pathogenetic mechanisms of DN by PA-induced oxidative stress. We conclude that activation of Nrf2-immunoproteasome signaling pathway by EPA/DHA plays a crucial role in abrogating the proteotoxic stress in DN. Antioxid. Redox Signal. 42, 751-766.
Collapse
Affiliation(s)
- Eugene Lee
- Renal Pathology Lab, Hankook Kidney and Diabetes Research Center, Seoul, Korea
| | - Jae-Hun Ahn
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Byeong-Cheol Kang
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Soon Lee
- Renal Pathology Lab, Hankook Kidney and Diabetes Research Center, Seoul, Korea
| |
Collapse
|
2
|
Agbozo WK, Solomon W, Lekpor CE, Erskine IJ, Oguljahan B, Bashi A, Harbuzariu A, Driss A, Adjei S, Paemka L, Ofori-Acquah SF, Stiles JK. Hydroxyurea Mitigates Heme-Induced Inflammation and Kidney Injury in Humanized Sickle Cell Mice. Int J Mol Sci 2025; 26:3214. [PMID: 40244015 PMCID: PMC11989777 DOI: 10.3390/ijms26073214] [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: 02/04/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Kidney disorders significantly contribute to morbidity and mortality in sickle cell disease (SCD). Acute kidney injury (AKI), a major risk factor for chronic kidney disease (CKD), often arises from intravascular hemolysis, where plasma cell-free heme drives AKI through inflammatory and oxidative stress mechanisms. Hydroxyurea (HU), a well-established SCD-modifying therapy, improves clinical outcomes, but its effects on systemic heme and inflammatory mediators of kidney injury remain underexplored. This study evaluated HU's impact on plasma heme, pro-inflammatory mediators, kidney injury, and renal histopathology in a sickle cell mouse model. Townes humanized sickle cell mice (HbSS) and non-sickle (HbAA) controls were treated with HU or vehicle for two weeks. HU significantly reduced total plasma heme, lactate dehydrogenase, and pro-inflammatory cytokines (CXCL10, VEGF-A, IFN-γ) in HbSS mice. HU reduced renal injury biomarkers (cystatin C, NGAL) and improved renal histopathology, evidenced by reduced vascular congestion, glomerulosclerosis, and tubular damage. Interestingly, HU did not alter the levels of kidney repair biomarkers (clusterin and EGF). These findings suggest that HU mitigates kidney injury by reducing the deleterious effects of circulating heme and inflammation, supporting its potential to slow or prevent progressive kidney injury in SCD.
Collapse
Affiliation(s)
- William Kwaku Agbozo
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (W.K.A.)
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, P.O. Box LG 25 Legon-Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, P.O. Box LG 25 Legon-Accra, Ghana
| | - Wesley Solomon
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (W.K.A.)
| | - Cecilia Elorm Lekpor
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (W.K.A.)
| | - Isaac Joe Erskine
- Department of Pathology, Korle-Bu Teaching Hospital, P.O. Box 77 Korle Bu-Accra, Ghana
| | - Babayewa Oguljahan
- Center for Laboratory Animal Resources, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Alaijah Bashi
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (W.K.A.)
| | | | - Adel Driss
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (W.K.A.)
| | - Samuel Adjei
- Department of Animal Experimentation, Noguchi Memorial Institute for Medical Research, University of Ghana, P.O. Box LG 581 Legon-Accra, Ghana
| | - Lily Paemka
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, P.O. Box LG 25 Legon-Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, P.O. Box LG 25 Legon-Accra, Ghana
| | - Solomon Fifii Ofori-Acquah
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (W.K.A.)
- West African Genetic Medicine Center (WAGMC), University of Ghana, P.O. Box LG 25 Legon-Accra, Ghana
| | - Jonathan K. Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (W.K.A.)
| |
Collapse
|
3
|
Li N, Song J, Yang Y, Huang X, Tian Y, Chen B, Lin L, Qin Z. Nrf2 protects against oxidative damage induced by hemoglobin in the liver of grass carp (Ctenopharyngodon idella). Biochim Biophys Acta Mol Basis Dis 2025; 1871:167600. [PMID: 39615659 DOI: 10.1016/j.bbadis.2024.167600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024]
Abstract
Hemoglobin (Hb) releases during hemorrhaging and causes oxidative damage, further exacerbates the development of multiple diseases. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates cellular defenses against toxic and oxidative challenges. However, the regulation mechanism of Nrf2 in Hb-induced oxidative stress remains unclear in teleost. To accomplish this goal, a hemolysis model was established by injecting grass carp with phenylalanine (PHZ), and the immunofluorescence analysis (IFA) and hematoxylin and eosin (H&E) staining revealed that PHZ-induced hemolysis caused Hb accumulation and hepatic vacuolization, resulted in tissue damage. Prussian blue, Sirius red, and Masson staining results revealed significant iron deposition and extensive collagen fiber accumulation in the liver. IFA and immunohistochemical analyses demonstrated that PHZ-induced hemolysis markedly increased the production of reactive oxygen species (ROS), malondialdehyde (MDA), and 4-hydroxynonenal (4-HNE). The quantitative real-time PCR (qRT-PCR) analysis data revealed that the PHZ-induced hemolysis also significantly upregulated the expression of antioxidant-related genes through activation of the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)/Nrf2 signaling pathway. To further explore the molecule regulation mechanism of PHZ-induced hemolysis, the RNA-seq analysis was performed, and the data revealed that the AMPK/Nrf2 and multiple programmed cell death pathways, including ferroptosis, autophagy, apoptosis, and necroptosis in PHZ injection groups were significant upregulated. In vitro, the hemin supplementation activated the expression of target genes in the AMPK/Nrf2 pathway detected by qRT-PCR. To further verify the regulation function of Nrf2, an Nrf2 activator (4OI) was supplemented, and the flow cytometer analysis results suggested that the Hb-induced cell damage was significantly attenuated. However, the supplementary of ML385 down-regulated the AMPK/Nrf2 pathway and aggravated the hemin induced cell death. In conclusion, these findings highlight the critical regulatory role of the AMPK/Nrf2 signaling pathway in protecting against Hb-induced oxidative damage in the liver of grass carp.
Collapse
Affiliation(s)
- Ningjing Li
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Jialing Song
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Yan Yang
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Xiaoman Huang
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Ye Tian
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Bing Chen
- Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
| | - Li Lin
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China.
| | - Zhendong Qin
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China.
| |
Collapse
|
4
|
Wang W, Chen J, Zhan L, Zou H, Wang L, Guo M, Gao H, Xu J, Wu W. Iron and ferroptosis in kidney disease: molecular and metabolic mechanisms. Front Immunol 2025; 16:1531577. [PMID: 39975561 PMCID: PMC11835690 DOI: 10.3389/fimmu.2025.1531577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Maintaining iron homeostasis is necessary for kidney functioning. There is more and more research indicating that kidney disease is often caused by iron imbalance. Over the past decade, ferroptosis' role in mediating the development and progression of renal disorders, such as acute kidney injury (renal ischemia-reperfusion injury, drug-induced acute kidney injury, severe acute pancreatitis induced acute kidney injury and sepsis-associated acute kidney injury), chronic kidney disease (diabetic nephropathy, renal fibrosis, autosomal dominant polycystic kidney disease) and renal cell carcinoma, has come into focus. Thus, knowing kidney iron metabolism and ferroptosis regulation may enhance disease therapy. In this review, we discuss the metabolic and molecular mechanisms of iron signaling and ferroptosis in kidney disease. We also explore the possible targets of ferroptosis in the therapy of renal illness, as well as their existing limitations and future strategies.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jingdi Chen
- Department of orthopedics, The Airborne Military Hospital, Wuhan, Hubei, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Handong Zou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mengmeng Guo
- The First Clinical College of Wuhan University, Wuhan, Hubei, China
| | - Hang Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
5
|
García-Caballero C, Guerrero-Hue M, Vallejo-Mudarra M, Palomino Antolin A, Decouty-Pérez C, Sánchez-Mendoza LM, Villalba JM, González-Reyes JA, Opazo-Rios L, Vázquez-Carballo C, Herencia C, Leiva-Cepas F, Cortegano I, Andrés BD, Egido J, Egea J, Moreno JA. Nox4 is involved in acute kidney injury associated to intravascular hemolysis. Free Radic Biol Med 2024; 225:430-444. [PMID: 39413979 DOI: 10.1016/j.freeradbiomed.2024.10.283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Massive intravascular hemolysis occurs not unfrequently in many clinical conditions. Breakdown of erythrocytes promotes the accumulation of heme-derivates in the kidney, increasing oxidative stress and cell death, thus promoting acute kidney injury (AKI). NADPH oxidase 4 (Nox4) is a major source of reactive oxygen species (ROS) in the kidney, however it is unknown the role of Nox4 in hemolysis and whether inhibition of this enzyme may protect from heme-mediated injury. To answer these questions, we elicited intravascular hemolysis in wild type and Nox4 knockout mice. We also evaluated whether nephrotoxic effects of heme may be reduced by using Nox4 siRNA and pharmacologic inhibition with GKT137831, a Nox4 inhibitor, both in vivo and in cultured renal cells. Our results showed that induction of massive hemolysis elicited AKI characterized by loss of renal function, morphological alterations of the tubular epithelium and podocytes, oxidative stress, inflammation, mitochondrial dysfunction, blockade of autophagy and cell death. These pathological effects were significantly prevented in Nox4-deficient mice and in animals treated with GKT137831. In vitro studies showed that Nox4 disruption by specific siRNAs or Nox4 inhibitors declined heme-mediated ROS production and cell death. Our data identify Nox4 as a key enzyme involved in intravascular hemolysis-induced AKI. Thus, Nox4 inhibition may be a potential therapeutic approach to prevent renal damage in patients with severe hemolytic crisis.
Collapse
Affiliation(s)
- Cristina García-Caballero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain.
| | - Melania Guerrero-Hue
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain.
| | - Mercedes Vallejo-Mudarra
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain.
| | - Alejandra Palomino Antolin
- Research Unit, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain.
| | - Celine Decouty-Pérez
- Research Unit, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain.
| | - Luz Marina Sánchez-Mendoza
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| | - José Manuel Villalba
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| | - José Antonio González-Reyes
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| | - Lucas Opazo-Rios
- Health Science Faculty, University of Las Américas, Concepción, Talcahuano, Chile.
| | - Cristina Vázquez-Carballo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Autonomous University Madrid, Spain.
| | - Carmen Herencia
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Autonomous University Madrid, Spain.
| | - Fernando Leiva-Cepas
- Departament of Morphological and Sociosanitary Sciences, Pathology Unit, Faculty of Medicine and Nurse, University of Cordoba/Pathology Unit, Hospital Universitario Reina Sofía, Cordoba, Spain.
| | - Isabel Cortegano
- Immunobiology Department, Carlos III Health Institute, Madrid, Spain.
| | - Belén de Andrés
- Immunobiology Department, Carlos III Health Institute, Madrid, Spain.
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Autonomous University Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| | - Javier Egea
- Research Unit, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006, Madrid, Spain.
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Campus of International Agri-Food Excellence, CeiA3, Cordoba, Spain.
| |
Collapse
|
6
|
Xie T, Yao L, Li X. Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases. Antioxidants (Basel) 2024; 13:659. [PMID: 38929098 PMCID: PMC11200795 DOI: 10.3390/antiox13060659] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Kidney diseases pose a significant global health issue, frequently resulting in the gradual decline of renal function and eventually leading to end-stage renal failure. Abnormal iron metabolism and oxidative stress-mediated cellular dysfunction facilitates the advancement of kidney diseases. Iron homeostasis is strictly regulated in the body, and disturbance in this regulatory system results in abnormal iron accumulation or deficiency, both of which are associated with the pathogenesis of kidney diseases. Iron overload promotes the production of reactive oxygen species (ROS) through the Fenton reaction, resulting in oxidative damage to cellular molecules and impaired cellular function. Increased oxidative stress can also influence iron metabolism through upregulation of iron regulatory proteins and altering the expression and activity of key iron transport and storage proteins. This creates a harmful cycle in which abnormal iron metabolism and oxidative stress perpetuate each other, ultimately contributing to the advancement of kidney diseases. The crosstalk of iron metabolism and oxidative stress involves multiple signaling pathways, such as hypoxia-inducible factor (HIF) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. This review delves into the functions and mechanisms of iron metabolism and oxidative stress, along with the intricate relationship between these two factors in the context of kidney diseases. Understanding the underlying mechanisms should help to identify potential therapeutic targets and develop novel and effective therapeutic strategies to combat the burden of kidney diseases.
Collapse
Affiliation(s)
- Tiancheng Xie
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
7
|
Labarque V, Okocha EC. Systematic Review of Genetic Modifiers Associated with the Development and/or Progression of Nephropathy in Patients with Sickle Cell Disease. Int J Mol Sci 2024; 25:5427. [PMID: 38791464 PMCID: PMC11121490 DOI: 10.3390/ijms25105427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/05/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Sickle cell nephropathy (SCN) is a common complication of sickle cell disease (SCD) that significantly contributes to morbidity and mortality. In addition to clinical and life-style factors, genetic variants influence this risk. We performed a systematic review, searching five databases. Studies evaluating the effect of genetic modifiers on SCN were eligible. Twenty-eight studies (fair-to-good quality) were included: one genome-wide association study, twenty-six case-control studies, and one article combining both approaches. APOL1 was significantly associated with albuminuria and hyperfiltration in children and with worse glomerular filtration in adults. On the other hand, alpha-thalassemia protected patients against albuminuria and hyperfiltration, while BCL11A variants were protective against albuminuria alone. The HMOX1 long GT-tandem repeat polymorphism led to a lower glomerular filtration rate. No modifiers for the risk of hyposthenuria were identified. A genome-wide association approach identified three new loci for proteinuria (CRYL1, VWF, and ADAMTS7) and nine loci were linked with eGFR (PKD1L2, TOR2A, CUBN, AGGF1, CYP4B1, CD163, LRP1B, linc02288, and FPGT-TNNI3K/TNNI3K). In conclusion, this systematic review supports the role of genetic modifiers in influencing the risk and progression of SCN. Incorporating and expanding this knowledge is crucial to improving the management and clinical outcomes of patients at risk.
Collapse
Affiliation(s)
- Veerle Labarque
- Department of Pediatric Hemato-Oncology, University Hospitals Leuven, 3000 Leuven, Belgium
- Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Emmanuel Chide Okocha
- Haematology Department, Faculty of Basic Clinical Sciences, College of Health Sciences, Nnamdi Azikiwe University, Nnewi PMB 5025, Anambra State, Nigeria
| |
Collapse
|
8
|
Li S, Han Q, Liu C, Wang Y, Liu F, Pan S, Zuo L, Gao D, Chen K, Feng Q, Liu Z, Liu D. Role of ferroptosis in chronic kidney disease. Cell Commun Signal 2024; 22:113. [PMID: 38347570 PMCID: PMC10860320 DOI: 10.1186/s12964-023-01422-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/03/2023] [Indexed: 02/15/2024] Open
Abstract
Chronic kidney disease (CKD) has historically been a significant global health concern, profoundly impacting both life and well-being. In the process of CKD, with the gradual loss of renal function, the incidence of various life-threatening complications, such as cardiovascular diseases, cerebrovascular accident, infection and stroke, is also increasing rapidly. Unfortunately, existing treatments exhibit limited ability to halt the progression of kidney injury in CKD, emphasizing the urgent need to delve into the precise molecular mechanisms governing the occurrence and development of CKD while identifying novel therapeutic targets. Renal fibrosis, a typical pathological feature of CKD, plays a pivotal role in disrupting normal renal structures and the loss of renal function. Ferroptosis is a recently discovered iron-dependent form of cell death characterized by lipid peroxide accumulation. Ferroptosis has emerged as a potential key player in various diseases and the initiation of organ fibrosis. Substantial evidence suggests that ferroptosis may significantly contribute to the intricate interplay between CKD and its progression. This review comprehensively outlines the intricate relationship between CKD and ferroptosis in terms of iron metabolism and lipid peroxidation, and discusses the current landscape of pharmacological research on ferroptosis, shedding light on promising avenues for intervention. It further illustrates recent breakthroughs in ferroptosis-related regulatory mechanisms implicated in the progression of CKD, thereby providing new insights for CKD treatment. Video Abstract.
Collapse
Affiliation(s)
- Shiyang Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Chang Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yixue Wang
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Fengxun Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Shaokang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Dan Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Kai Chen
- Kaifeng Renmin Hospital, Kaifeng, 475000, Henan, People's Republic of China
| | - Qi Feng
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Dongwei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
9
|
Li J, Li L, Zhang Z, Chen P, Shu H, Yang C, Chu Y, Liu J. Ferroptosis: an important player in the inflammatory response in diabetic nephropathy. Front Immunol 2023; 14:1294317. [PMID: 38111578 PMCID: PMC10725962 DOI: 10.3389/fimmu.2023.1294317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/15/2023] [Indexed: 12/20/2023] Open
Abstract
Diabetic nephropathy (DN) is a chronic inflammatory disease that affects millions of diabetic patients worldwide. The key to treating of DN is early diagnosis and prevention. Once the patient enters the clinical proteinuria stage, renal damage is difficult to reverse. Therefore, developing early treatment methods is critical. DN pathogenesis results from various factors, among which the immune response and inflammation play major roles. Ferroptosis is a newly discovered type of programmed cell death characterized by iron-dependent lipid peroxidation and excessive ROS production. Recent studies have demonstrated that inflammation activation is closely related to the occurrence and development of ferroptosis. Moreover, hyperglycemia induces iron overload, lipid peroxidation, oxidative stress, inflammation, and renal fibrosis, all of which are related to DN pathogenesis, indicating that ferroptosis plays a key role in the development of DN. Therefore, this review focuses on the regulatory mechanisms of ferroptosis, and the mutual regulatory processes involved in the occurrence and development of DN and inflammation. By discussing and analyzing the relationship between ferroptosis and inflammation in the occurrence and development of DN, we can deepen our understanding of DN pathogenesis and develop new therapeutics targeting ferroptosis or inflammation-related regulatory mechanisms for patients with DN.
Collapse
Affiliation(s)
- Jialing Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Luxin Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
- School of First Clinical Medical College, Mudanjiang Medical University, Mudanjiang, China
| | - Peijian Chen
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Haiying Shu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Can Yang
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Yanhui Chu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Jieting Liu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
10
|
Zand L, Fervenza FC, Coppo R. Microscopic hematuria as a risk factor for IgAN progression: considering this biomarker in selecting and monitoring patients. Clin Kidney J 2023; 16:ii19-ii27. [PMID: 38053974 PMCID: PMC10695511 DOI: 10.1093/ckj/sfad232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 12/07/2023] Open
Abstract
Hematuria-either macroscopic hematuria or asymptomatic microscopic hematuria-is a clinical feature typical but not specific for immunoglobulin A nephropathy (IgAN). The only biomarker supported by the Kidney Disease: Improving Global Outcomes group as a predictor of progression, identifying patients needing treatment, is proteinuria >1 g/day persistent despite maximized supportive care. However, proteinuria can occur in the setting of active glomerulonephritis or secondary to sclerotic renal lesions. Microscopic hematuria is observed in experimental models of IgAN after IgA-IgG immunocomplex deposition, activation of inflammation and complement pathways. Oxidative damage, triggered by hemoglobin release, is thought to contribute to the development of proteinuria and progression. Despite being a clinical hallmark of IgAN and having a rational relationship with its pathophysiology, the value of microscopic hematuria in assessing activity and predicting outcomes in patients with IgAN is still debated. This was partly due to a lack of standardization and day-to-day variability of microhematuria, which discouraged the inclusion of microhematuria in large multicenter studies. More recently, several studies from Asia, Europe and the USA have highlighted the importance of microhematuria assessment over longitudinal follow-up, using a systematic approach with either experienced personnel or automated techniques. We report lights and shadows of microhematuria evaluation in IgAN, looking for evidence for a more consistent consensus on its value as a marker of clinical and histological activity, risk assessment and prediction of treatment response. We propose that hematuria should be included as part of the clinical decision-making process when considering when to use immunosuppressive therapy and as part of criteria for enrollment into clinical trials to test drugs targeting the inflammatory reaction elicited by immune pathway activation in IgAN.
Collapse
Affiliation(s)
- Ladan Zand
- Division of Nephrology and Hypertension. Mayo Clinic. Rochester, MN, USA
| | | | - Rosanna Coppo
- Fondazione Ricerca Molinette, Regina Margherita Hospital, Turin, Italy
| |
Collapse
|
11
|
García-Giménez J, Córdoba-David G, Rayego-Mateos S, Cannata-Ortiz P, Carrasco S, Ruiz-Ortega M, Fernandez-Fernandez B, Ortiz A, Ramos AM. STING1 deficiency ameliorates immune-mediated crescentic glomerulonephritis in mice. J Pathol 2023; 261:309-322. [PMID: 37650295 DOI: 10.1002/path.6177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Rapidly progressive/crescentic glomerulonephritis (RPGN/CGN) involves the formation of glomerular crescents by maladaptive differentiation of parietal epithelial cells that leads to rapid loss of renal function. The molecular mechanisms of crescent formation are poorly understood. Therefore, new insights into molecular mechanisms could identify alternative therapeutic targets for RPGN/CGN. Analysis of kidney biopsies from patients with RPGN revealed increased interstitial, glomerular, and tubular expression of STING1, an accessory protein of the c-GAS-dependent DNA-sensing pathway, which was also observed in murine nephrotoxic nephritis induced by an anti-GBM antibody. STING1 was expressed by key cell types involved in RPGN and crescent formation such as glomerular parietal epithelial cells, and tubular cells as well as by inflammation accessory cells. In functional in vivo studies, Sting1-/- mice with nephrotoxic nephritis had lower kidney cytokine expression, milder kidney infiltration by innate and adaptive immune cells, and decreased disease severity. Pharmacological STING1 inhibition mirrored these findings. Direct STING1 agonism in parietal and tubular cells activated the NF-κB-dependent cytokine response and the interferon-induced genes (ISGs) program. These responses were also triggered in a STING1-dependent manner by the pro-inflammatory cytokine TWEAK. These results identify STING1 activation as a pathological mechanism in RPGN/CGN and TWEAK as an activator of STING1. Pharmacological strategies targeting STING1, or upstream regulators may therefore be potential alternatives to treat RPGN. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jorge García-Giménez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Gina Córdoba-David
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Pablo Cannata-Ortiz
- Department of Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Carrasco
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Pharmacology, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Adrián M Ramos
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
12
|
Sevillano AM, Caravaca-Fontán F, Cordero Garcia-Galan L, Fernandez-Juarez G, Lopez-Revuelta K, Guzmán DA, Martín-Reyes G, Quintana LF, Rodas LM, Sanchez de la Nieta MD, Rabasco C, Espinosa M, Diaz-Encarnación M, San Miguel L, Barrios C, Rodriguez E, Garcia P, Valera A, Peña JK, Shabaka A, Velo M, Sierra M, Gonzalez F, Fernandez-Reyes MJ, Heras M, Delgado P, Gutierrez E, Moreno JA, Praga M. Effect of Immunosuppressive Treatments on Kidney Outcomes After Gross Hematuria-Related Acute Kidney Injury in Older Patients With IgA Nephropathy. Kidney Int Rep 2023; 8:1596-1604. [PMID: 37547537 PMCID: PMC10403672 DOI: 10.1016/j.ekir.2023.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 08/08/2023] Open
Abstract
INTRODUCTION Macroscopic hematuria (MH) bouts, frequently accompanied by acute kidney injury (AKI-MH) are one of the most common presentations of IgA nephropathy (IgAN) in the elderly. Immunosuppressive therapies are used in clinical practice; however, no studies have analyzed their efficacy on kidney outcomes. METHODS This is a retrospective, multicenter study of a cohort of patients aged ≥50 years with biopsy-proven IgAN presenting with AKI-MH. Outcomes were complete, partial, or no recovery of kidney function at 1 year after AKI-MH, and kidney survival at 1, 2, and 5 years. Propensity score matching (PSM) analysis was applied to balance baseline differences between patients treated with immunosuppression and those not treated with immunosuppression. RESULTS The study group consisted of 91 patients with a mean age of 65 ± 15 years, with a mean follow-up of 59 ± 36 months. Intratubular red blood cell (RBC) casts and acute tubular necrosis were found in all kidney biopsies. The frequency of endocapillary hypercellularity and crescents were low. Immunosuppressive therapies (corticosteroids alone or combined with mycophenolate mofetil or cyclophosphamide) were prescribed in 52 (57%) patients, whereas 39 (43%) received conservative treatment. There were no significant differences in the proportion of patients with complete, partial, or no recovery of kidney function at 1 year between patients treated with immunosuppression and those not treated with immunosuppression (29% vs. 36%, 30.8% vs. 20.5% and 40.4 % vs. 43.6%, respectively). Kidney survival at 1, 3, and 5 years was similar among treated and untreated patients (85% vs. 81%, 77% vs. 76% and 72% vs. 66%, respectively). Despite the PSM analysis, no significant differences were observed in kidney survival between the two groups. Fourteen patients (27%) treated with immunosuppression had serious adverse events. CONCLUSIONS Immunosuppressive treatments do not modify the unfavorable prognosis of patients with IgAN who are aged ≥50 years presenting with AKI-MH, and are frequently associated with severe complications.
Collapse
Affiliation(s)
- Angel M. Sevillano
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Caravaca-Fontán
- Department of Nephrology, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Gema Fernandez-Juarez
- Department of Nephrology, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Katia Lopez-Revuelta
- Department of Nephrology, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Diomaris A. Guzmán
- Department of Nephrology, Hospital Regional Universitario de Málaga, Malaga, Spain
| | | | - Luis F. Quintana
- Department of Nephrology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Lida M. Rodas
- Department of Nephrology, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Cristina Rabasco
- Department of Nephrology, Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Mario Espinosa
- Department of Nephrology, Hospital Universitario Reina Sofia, Cordoba, Spain
| | | | - Luz San Miguel
- Department of Nephrology, Fundación Puigvert, Barcelona, Spain
| | - Clara Barrios
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Eva Rodriguez
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Patricia Garcia
- Department of Nephrology, Hospital Virgen de la Victoria, Malaga, Spain
| | - Alfonso Valera
- Department of Nephrology, Hospital Virgen de la Victoria, Malaga, Spain
| | - Jessy-Korina Peña
- Department of Nephrology, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain
| | - Amir Shabaka
- Department of Nephrology, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Mercedes Velo
- Department of Nephrology, Hospital Clínico San Carlos, Madrid, Spain
| | - Milagros Sierra
- Department of Nephrology, Hospital San Pedro, Logroño, Spain
| | - Fayna Gonzalez
- Department of Nephrology, Hospital Universitario Dr. Negrin, Gran Canaria, Spain
| | | | - Manuel Heras
- Department of Nephrology, Hospital General de Segovia, Segovia, Spain
| | - Patricia Delgado
- Department of Nephrology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Eduardo Gutierrez
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Juan Antonio Moreno
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), UGC Nefrología, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Manuel Praga
- Department of Nephrology, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Medicine Department, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
13
|
Williams MC, Zhang X, Baek JH, D’Agnillo F. Renal glomerular and tubular responses to glutaraldehyde- polymerized human hemoglobin. Front Med (Lausanne) 2023; 10:1158359. [PMID: 37384048 PMCID: PMC10293615 DOI: 10.3389/fmed.2023.1158359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/30/2023] [Indexed: 06/30/2023] Open
Abstract
Hemoglobin-based oxygen carriers (HBOCs) are being developed as oxygen and volume replacement therapeutics, however, their molecular and cellular effects on the vasculature and different organ systems are not fully defined. Using a guinea pig transfusion model, we examined the renal glomerular and tubular responses to PolyHeme, a highly characterized glutaraldehyde-polymerized human hemoglobin with low tetrameric hemoglobin content. PolyHeme-infused animals showed no major changes in glomerular histology or loss of specific markers of glomerular podocytes (Wilms tumor 1 protein, podocin, and podocalyxin) or endothelial cells (ETS-related gene and claudin-5) after 4, 24, and 72 h. Relative to sham controls, PolyHeme-infused animals also showed similar expression and subcellular distribution of N-cadherin and E-cadherin, two key epithelial junctional proteins of proximal and distal tubules, respectively. In terms of heme catabolism and iron-handling responses, PolyHeme induced a moderate but transient expression of heme oxygenase-1 in proximal tubular epithelium and tubulointerstitial macrophages that was accompanied by increased iron deposition in tubular epithelium. Contrary to previous findings with other modified or acellular hemoglobins, the present data show that PolyHeme does not disrupt the junctional integrity of the renal glomerulus and tubular epithelium, and triggers moderate activation of heme catabolic and iron sequestration systems likely as part of a renal adaptive response.
Collapse
|
14
|
Zhai H, Ni L, Wu X. The roles of heme oxygenase-1 in renal disease. FRONTIERS IN NEPHROLOGY 2023; 3:1156346. [PMID: 37675385 PMCID: PMC10479750 DOI: 10.3389/fneph.2023.1156346] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/17/2023] [Indexed: 09/08/2023]
Abstract
Heme oxygenase (HO), a heat shock protein containing hemoglobin, is an important enzyme in heme catabolism. It is involved in cell homeostasis and has anti-inflammatory, antioxidant, anti-apoptosis, immunomodulation, and other functions. It is expressed at a modest level in most normal tissues. When the body suffers from ischemia hypoxia, injury, toxins, and other nociceptive stimuli, the expression increases, which can transform the oxidative microenvironment into an antioxidant environment to promote tissue recovery from damage. In recent years, research has continued to verify its value in a variety of human bodily systems. It is also regarded as a key target for the treatment of numerous disorders. With the advancement of studies, its significance in renal disease has gained increasing attention. It is thought to have a significant protective function in preventing acute kidney injury and delaying the progression of chronic renal diseases. Its protective mechanisms include anti-inflammatory, antioxidant, cell cycle regulation, apoptosis inhibition, hemodynamic regulation, and other aspects, which have been demonstrated in diverse animal models. Furthermore, as a protective factor, its potential therapeutic efficacy in renal disease has recently become a hot area of research. Although a large number of preclinical trials have confirmed its therapeutic potential in reducing kidney injury, due to the problems and side effects of HO-1 induction therapy, its efficacy and safety in clinical application need to be further explored. In this review, we summarize the current state of research on the mechanism, location, and treatment of HO and its relationship with various renal diseases.
Collapse
Affiliation(s)
- Hongfu Zhai
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lihua Ni
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaoyan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of General Practice, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Wang W, Pan C, Lv M, Ruan Q, Chen W, Shafique L, Parveen S, Liang Z, Ma H, Luo X. Effect of hemoglobin on Nile tilapia (Oreochromis niloticus) kidney (NTK) cell line damage. FISH & SHELLFISH IMMUNOLOGY 2022; 131:637-645. [PMID: 36272521 DOI: 10.1016/j.fsi.2022.10.029] [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: 06/07/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
Bacteria or viral outbreaks can cause tilapia hemorrhage, ensuring considerable volume of hemoglobin (Hb) into the tissue. However, the hemoglobin toxicity on tissue and high doses also effect on tissue this phenomena is still under consideration. Therefore, current study exploited Nile tilapia kidney (NTK) cells to deeply expose the toxic effect of Hb on NTK cells. Toxicity of Hb on NTK cells was determined in terms of cells growth, expression of iron metabolism and inflammation-related genes, consequently examined antioxidant-related enzymes genes expression, intracellular iron and reactive oxygen species (ROS) contents, and apoptosis-related genes expression. The results showed that Hb and heme significantly inhibited NTK cells growth and up-regulated iron metabolism-related genes expression in different degrees. The Hb and heme activated the expression of pro-inflammatory cytokines (TNF-α, tumor necrosis factor-α; IL-1β, interleukin 1β; IL-6, interleukin 6), the anti-inflammatory factor (IL-10, interleukin 10) and the chemotactic factors (IL-4, interleukin 4; IL-8, interleukin 8) through NF-κB pathway, meanwhile activated the expression of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px). Moreover, the Hb significantly increased intracellular iron and ROS contents while the expression of apoptosis-related genes was significantly activated by both Hb and heme. Current investigation suggested that high oxidative activity of Hb could activate iron metabolism- and inflammation-related genes expression, and increase intracellular iron and ROS levels, lead to up-regulated the expression of apoptosis genes in NTK cells.
Collapse
Affiliation(s)
- Weisheng Wang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, China.
| | - Chuanyan Pan
- Center of Aquaculture and Processing Technology, Guangxi Academy of Fishery Science, Nanning, 530021, Guangxi, China
| | - Min Lv
- Center of Aquaculture and Processing Technology, Guangxi Academy of Fishery Science, Nanning, 530021, Guangxi, China; The Food Engineering and Technology Center, Guangxi Xiaoyanren Biotechnology Co., Ltd, Nanning, 530017, Guangxi, China
| | - Qiufeng Ruan
- Center of Aquaculture and Processing Technology, Guangxi Academy of Fishery Science, Nanning, 530021, Guangxi, China
| | - Weijie Chen
- Center of Aquaculture and Processing Technology, Guangxi Academy of Fishery Science, Nanning, 530021, Guangxi, China
| | - Laiba Shafique
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Shakeela Parveen
- Department of Zoology, Government Sadiq College Women University, Bahawalpur, Pakistan
| | - Zheng Liang
- Center of Aquaculture and Processing Technology, Guangxi Academy of Fishery Science, Nanning, 530021, Guangxi, China
| | - Huawei Ma
- Center of Aquaculture and Processing Technology, Guangxi Academy of Fishery Science, Nanning, 530021, Guangxi, China
| | - Xu Luo
- Center of Aquaculture and Processing Technology, Guangxi Academy of Fishery Science, Nanning, 530021, Guangxi, China
| |
Collapse
|
16
|
Nath KA, Singh RD, Croatt AJ, Adams CM. Heme Proteins and Kidney Injury: Beyond Rhabdomyolysis. KIDNEY360 2022; 3:1969-1979. [PMID: 36514409 PMCID: PMC9717624 DOI: 10.34067/kid.0005442022] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022]
Abstract
Heme proteins, the stuff of life, represent an ingenious biologic strategy that capitalizes on the biochemical versatility of heme, and yet is one that avoids the inherent risks to cellular vitality posed by unfettered and promiscuously reactive heme. Heme proteins, however, may be a double-edged sword because they can damage the kidney in certain settings. Although such injury is often viewed mainly within the context of rhabdomyolysis and the nephrotoxicity of myoglobin, an increasing literature now attests to the fact that involvement of heme proteins in renal injury ranges well beyond the confines of this single disease (and its analog, hemolysis); indeed, through the release of the defining heme motif, destabilization of intracellular heme proteins may be a common pathway for acute kidney injury, in general, and irrespective of the underlying insult. This brief review outlines current understanding regarding processes underlying such heme protein-induced acute kidney injury (AKI) and chronic kidney disease (CKD). Topics covered include, among others, the basis for renal injury after the exposure of the kidney to and its incorporation of myoglobin and hemoglobin; auto-oxidation of myoglobin and hemoglobin; destabilization of heme proteins and the release of heme; heme/iron/oxidant pathways of renal injury; generation of reactive oxygen species and reactive nitrogen species by NOX, iNOS, and myeloperoxidase; and the role of circulating cell-free hemoglobin in AKI and CKD. Also covered are the characteristics of the kidney that render this organ uniquely vulnerable to injury after myolysis and hemolysis, and pathobiologic effects emanating from free, labile heme. Mechanisms that defend against the toxicity of heme proteins are discussed, and the review concludes by outlining the therapeutic strategies that have arisen from current understanding of mechanisms of renal injury caused by heme proteins and how such mechanisms may be interrupted.
Collapse
Affiliation(s)
- Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher M. Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic Rochester, Minnesota
| |
Collapse
|
17
|
Weng M, Lin J, Chen Y, Zhang X, Zou Z, Chen Y, Cui J, Fu B, Li G, Chen C, Wan J. Time-Averaged Hematuria as a Prognostic Indicator of Renal Outcome in Patients with IgA Nephropathy. J Clin Med 2022; 11:jcm11226785. [PMID: 36431262 PMCID: PMC9694958 DOI: 10.3390/jcm11226785] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
We aim to investigate the association of time-averaged hematuria (TA-hematuria) with the progression of IgA nephropathy (IgAN). Based on TA-hematuria during follow-up, 152 patients with IgAN were divided into a hematuria remission group (≤28 red blood cells [RBCs]/μL) and a persistent hematuria group (>28 RBCs/μL). The persistent hematuria group had a higher percentage of patients with macroscopic hematuria, lower levels of hemoglobin and TA-serum albumin, and more severe renal pathologic lesions. The composite endpoint is defined as a doubling of the baseline SCr level (D-SCr), or the presence of ESRD. During the mean follow-up of 58.08 ± 23.51 months, 15 patients (9.9%) reached the primary outcome of ESRD and 19 patients (12.5%) reached the combined renal endpoint. Kaplan-Meier analysis showed that the persistent hematuria group had a lower renal survival rate. The persistent hematuria patients who were incorporated with proteinuria (≥1.0 g/day) and low TA-serum albumin (<40 g/L) had the worst renal outcomes. Multivariate Cox regression indicated that TA-hematuria (hazard ratio [HR] = 0.004, 95% CI: 0.001, 0.008; p = 0.010) was independently associated with the progression of IgAN. Receiver operating characteristic analysis indicated the optimal TA-hematuria cutoff value for predicting the progression of IgAN was 201.21 RBCs/μL in females and 37.25 RBCs/μL in males.
Collapse
Affiliation(s)
- Mengjie Weng
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jiaqun Lin
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Yumei Chen
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xiaohong Zhang
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Zhenhuan Zou
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Yi Chen
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jiong Cui
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Binbin Fu
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Guifen Li
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Caiming Chen
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Correspondence:
| |
Collapse
|
18
|
Adebayo OC, Van den Heuvel LP, Olowu WA, Levtchenko EN, Labarque V. Sickle cell nephropathy: insights into the pediatric population. Pediatr Nephrol 2022; 37:1231-1243. [PMID: 34050806 DOI: 10.1007/s00467-021-05126-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/10/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
Abstract
The life expectancy of individuals with sickle cell disease has increased over the years, majorly due to an overall improvement in diagnosis and medical care. Nevertheless, this improved longevity has resulted in an increased prevalence of chronic complications such as sickle cell nephropathy (SCN), which poses a challenge to the medical care of the patient, shortening the lifespan of patients by 20-30 years. Clinical presentation of SCN is age-dependent, with kidney dysfunction slowly beginning to develop from childhood, progressing to chronic kidney disease and kidney failure during the third and fourth decades of life. This review explores the epidemiology, pathology, pathophysiology, clinical presentation, and management of SCN by focusing on the pediatric population. It also discusses the factors that can modify SCN susceptibility.
Collapse
Affiliation(s)
- Oyindamola C Adebayo
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium.,Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lambertus P Van den Heuvel
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Pediatric Nephrology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Wasiu A Olowu
- Pediatric Nephrology and Hypertension Unit, Obafemi Awolowo University Teaching Hospitals Complex, Ile-Ife, Nigeria
| | - Elena N Levtchenko
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium. .,Department of Pediatric Nephrology, University Hospital Leuven, Herestraat 49, Bus 817, 3000, Leuven, Belgium.
| | - Veerle Labarque
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Pediatric Hematology, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Zhang Y, Mou Y, Zhang J, Suo C, Zhou H, Gu M, Wang Z, Tan R. Therapeutic Implications of Ferroptosis in Renal Fibrosis. Front Mol Biosci 2022; 9:890766. [PMID: 35655759 PMCID: PMC9152458 DOI: 10.3389/fmolb.2022.890766] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/12/2022] [Indexed: 12/13/2022] Open
Abstract
Renal fibrosis is a common feature of chronic kidney disease (CKD), and can lead to the destruction of normal renal structure and loss of kidney function. Little progress has been made in reversing fibrosis in recent years. Ferroptosis is more immunogenic than apoptosis due to the release and activation of damage-related molecular patterns (DAMPs) signals. In this paper, the relationship between renal fibrosis and ferroptosis was reviewed from the perspective of iron metabolism and lipid peroxidation, and some pharmaceuticals or chemicals associated with both ferroptosis and renal fibrosis were summarized. Other programmed cell death and ferroptosis in renal fibrosis were also firstly reviewed for comparison and further investigation.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhua Mou
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jianjian Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanjian Suo
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai Zhou
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Gu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Ruoyun Tan,
| |
Collapse
|
20
|
An evaluation of the roles of hematuria and uric acid in defining the prognosis of patients with IgA nephropathy. Pediatr Nephrol 2022; 37:947-958. [PMID: 33982147 DOI: 10.1007/s00467-021-05092-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/01/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
In recent years, many significant advances have been made in determining which clinical manifestations and pathologic lesions can provide prognostic information for patients with IgA nephropathy (IgAN). However, some important questions remain, including the long-term consequences of hematuria, both macroscopic (MH) and microscopic (mH), in patients with IgAN. The importance of distinguishing patients who have a single episode of MH of long duration from those with recurrent episodes of short duration and the prognostic importance of the episodes of acute kidney injury (AKI) that sometimes accompany episodic MH will be discussed. Studies that have evaluated the mechanisms that may be responsible for recurrent MH and the toxic effects of red blood cells (RBCs), or their constituents, on kidney tubules will also be addressed. In the last section, I will review the evidence that hyperuricemia (HU) may be a significant independent risk factor for progressive kidney disease in patients with IgAN.
Collapse
|
21
|
Saurage E, Davis PR, Meek R, Pollock DM, Kasztan M. Endothelin A receptor antagonist attenuated renal iron accumulation in iron overload heme oxygenase-1 knockout mice. Can J Physiol Pharmacol 2022; 100:637-650. [PMID: 35413222 PMCID: PMC10164438 DOI: 10.1139/cjpp-2022-0038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Progressive iron accumulation and renal impairment are prominent in both patients and mouse models of sickle cell disease (SCD). Endothelin A receptor (ETA) antagonism prevents this iron accumulation phenotype and reduces renal iron deposition in proximal tubules of SCD mice. To better understand the mechanisms of iron metabolism in the kidney and the role of ETA receptor in iron chelation and transport, we studied renal iron handling in a non-sickle cell iron overload model, heme oxygenase-1 (Hmox-1-/-) knockout mice. We found that Hmox-1-/- mice had elevated plasma endothelin-1 (ET-1), cortical ET-1 mRNA expression, and renal iron content compared to Hmox-1+/+ controls. The ETA receptor antagonist, ambrisentan, attenuated renal iron deposition, without any changes to anemia status in Hmox-1-/- mice. This was accompanied by reduced urinary iron excretion. Finally, ambrisentan had an important iron recycling effect by increasing expression of cellular iron exporter, ferroportin-1 (FPN-1) and circulating total iron levels in Hmox-1-/- mice. These findings suggest the ET-1/ETA signaling pathway contributes to in renal iron trafficking in a murine model of iron overload.
Collapse
Affiliation(s)
- Elizabeth Saurage
- The University of Alabama at Birmingham School of Medicine, 9967, Medicine, Division of Nephrology, Birmingham, Alabama, United States;
| | - Parker Ross Davis
- The University of Alabama at Birmingham Department of Medicine, 164494, Medicine, Division of Nephrology, Birmingham, Alabama, United States;
| | - Rachel Meek
- The University of Alabama at Birmingham School of Medicine, 9967, Medicine, Devision of Nephrology, Birmingham, Alabama, United States;
| | - David M Pollock
- The University of Alabama at Birmingham Department of Medicine, 164494, Medicine, Division of Nephrology, Birmingham, Alabama, United States;
| | - Malgorzata Kasztan
- The University of Alabama at Birmingham School of Medicine, 9967, Pediatrics, Division of Hematology-Oncology, Birmingham, Alabama, United States;
| |
Collapse
|
22
|
Carriazo S, Villalvazo P, Ortiz A. More on the invisibility of chronic kidney disease… and counting. Clin Kidney J 2022; 15:388-392. [PMID: 35198154 PMCID: PMC8690216 DOI: 10.1093/ckj/sfab240] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Indexed: 12/20/2022] Open
Abstract
Lack of awareness of a diagnosis of chronic kidney disease (CKD) in patients and physicians is a major contributor to fueling the CKD pandemic by also making it invisible to researchers and health authorities. This is an urgent matter to tackle if dire predictions of future CKD burden are to be addressed. CKD is set to become the fifth-leading global cause of death by 2040 and the second-leading cause of death before the end of the century in some countries with long life expectancy. Coronavirus disease 2019 (COVID-19) illustrated this invisibility: only after the summer of 2020 did it become clear that CKD was a major driver of COVID-19 mortality, both in terms of prevalence as a risk factor and of the risk conferred for lethal COVID-19. However, by that time the damage was done: news outlets and scientific publications continued to list diabetes and hypertension, but not CKD, as major risk factors for severe COVID-19. In a shocking recent example from Sweden, CKD was found to be diagnosed in just 23% of 57 880 persons who fulfilled diagnostic criteria for CKD. In the very same large cohort, diabetes or cancer were diagnosed in 29% of persons, hypertension in 82%, cardiovascular disease in 39% and heart failure in 28%. Thus, from the point of view of physicians, patients and health authorities, CKD was the least common comorbidity in persons with CKD, ranking sixth, after other better-known conditions. One of the consequences of this lack of awareness was that nephrotoxic medications were more commonly prescribed in patients with CKD who did not have a diagnosis of CKD. Low awareness of CKD may also fuel concepts such as the high prevalence of hypertensive nephropathy when CKD is diagnosed after the better-known condition of hypertension.
Collapse
Affiliation(s)
- Sol Carriazo
- Instituto de Investigación Sanitaria Fundacion Jimenez Diaz, Madrid, Spain
- Red de Investigación Renal (REDINREN), Madrid, Spain
| | | | - Alberto Ortiz
- Instituto de Investigación Sanitaria Fundacion Jimenez Diaz, Madrid, Spain
- Red de Investigación Renal (REDINREN), Madrid, Spain
| |
Collapse
|
23
|
Qin Z, Yang M, Lu Z, Babu VS, Li Y, Shi F, Zhan F, Liu C, Li J, Lin L. The Oxidative Injury of Extracellular Hemoglobin Is Associated With Reactive Oxygen Species Generation of Grass Carp (Ctenopharyngodon idella). Front Immunol 2022; 13:843662. [PMID: 35265088 PMCID: PMC8899113 DOI: 10.3389/fimmu.2022.843662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Intravascular hemolysis is a fundamental feature of hemorrhagic venereal infection or tissue and releases the endogenous damage-associated molecular pattern hemoglobin (Hb) into the plasma or tissues, which results in systemic inflammation, vasomotor dysfunction, thrombophilia, and proliferative vasculopathy. However, how the cytotoxic Hb affects the tissues of grass carp remains unclear. Here, we established a hemolysis model in grass carp by injecting phenylhydrazine (PHZ). The data revealed that the PHZ-induced hemolysis increased the content of Hb and activated the antioxidant system in plasma. The histopathology analysis data showed that the PHZ-induced hemolysis increased the accumulation of Hb and iron both in the head and middle kidney. The results of quantitative real-time PCR (qRT-PCR) detection suggested that the hemolysis upregulated the expressions of iron metabolism-related genes. In addition, the immunofluorescence and immunohistochemistry data revealed that the hemolysis caused an obvious deposition of collagen fiber, malondialdehyde (MDA), and 4-hydroxynonenal (4-HNE) accumulation and increased the content of oxidative-related enzymes such as β-galactosidase (β-GAL), lipid peroxide (LPO), and MDA in both the head and middle kidney. Furthermore, the PHZ-induced hemolysis significantly increased the production of reactive oxygen species (ROS), which resulted in apoptosis and modulated the expressions of cytokine-related genes. Taken together, excess of Hb released from hemolysis caused tissue oxidative damage, which may be associated with ROS and inflammation generation.
Collapse
Affiliation(s)
- Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Minxuan Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Zhijie Lu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - V. Sarath Babu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Yanan Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Fei Shi
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Fanbin Zhan
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Chun Liu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Jun Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- School of Sciences and Medicine, Lake Superior State University, Sault Ste. Marie, MI, United States
- *Correspondence: Li Lin, ; Jun Li,
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- *Correspondence: Li Lin, ; Jun Li,
| |
Collapse
|
24
|
Molecular Mechanisms of Kidney Injury and Repair. Int J Mol Sci 2022; 23:ijms23031542. [PMID: 35163470 PMCID: PMC8835923 DOI: 10.3390/ijms23031542] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) will become the fifth global cause of death by 2040, thus emphasizing the need to better understand the molecular mechanisms of damage and regeneration in the kidney. CKD predisposes to acute kidney injury (AKI) which, in turn, promotes CKD progression. This implies that CKD or the AKI-to-CKD transition are associated with dysfunctional kidney repair mechanisms. Current therapeutic options slow CKD progression but fail to treat or accelerate recovery from AKI and are unable to promote kidney regeneration. Unraveling the cellular and molecular mechanisms involved in kidney injury and repair, including the failure of this process, may provide novel biomarkers and therapeutic tools. We now review the contribution of different molecular and cellular events to the AKI-to-CKD transition, focusing on the role of macrophages in kidney injury, the different forms of regulated cell death and necroinflammation, cellular senescence and the senescence-associated secretory phenotype (SAPS), polyploidization, and podocyte injury and activation of parietal epithelial cells. Next, we discuss key contributors to repair of kidney injury and opportunities for their therapeutic manipulation, with a focus on resident renal progenitor cells, stem cells and their reparative secretome, certain macrophage subphenotypes within the M2 phenotype and senescent cell clearance.
Collapse
|
25
|
Lu J, Zhao Y, Liu M, Lu J, Guan S. Toward improved human health: Nrf2 plays a critical role in regulating ferroptosis. Food Funct 2021; 12:9583-9606. [PMID: 34542140 DOI: 10.1039/d1fo01036k] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ferroptosis is a recently defined type of regulated cell death caused by an excess iron-dependent accumulation of lipid peroxides and is morphologically and biochemically distinct from other types of cell death. Notably, Nrf2 is identified to exquisitely modulate ferroptosis due to its ability to target a host of ferroptosis cascade genes, which places Nrf2 in the pivotal position of ferroptosis. This paper reviews the regulation effect of Nrf2 on ferroptosis, different activation mechanisms of Nrf2 as well as the relevance of the Nrf2-ferroptosis axis in diseases, and finally summarizes foods with beneficial effects in ferroptosis via the Nrf2 pathway and aims to serve as a reference for follow-up studies of food functions related to Nrf2, ferroptosis, and human health.
Collapse
Affiliation(s)
- Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China. .,Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Yanan Zhao
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China.
| | - Meitong Liu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China.
| | - Jianing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China.
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China. .,Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
26
|
Adebayo OC, Betukumesu DK, Nkoy AB, Adesoji OM, Ekulu PM, Van den Heuvel LP, Levtchenko EN, Labarque V. Clinical and genetic factors are associated with kidney complications in African children with sickle cell anaemia. Br J Haematol 2021; 196:204-214. [PMID: 34545573 DOI: 10.1111/bjh.17832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/19/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022]
Abstract
Clinical and genetic factors have been reported as influencing the development of sickle cell nephropathy (SCN). However, such data remain limited in the paediatric population. In this cross-sectional study, we enrolled 361 sickle cell disease children from the Democratic Republic of Congo. Participants were genotyped for the beta (β)-globin gene, apolipoprotein L1 (APOL1) risk variants, and haem oxygenase-1 (HMOX1) GT-dinucleotide repeats. As markers of kidney damage, albuminuria, hyperfiltration and decreased estimated glomerular filtration with creatinine (eGFRcr) were measured. An association of independent clinical and genetic factors with these markers of kidney damage were assessed via regression analysis. Genetic sequencing confirmed sickle cell anaemia in 326 participants. Albuminuria, hyperfiltration and decreased eGFRcr were present in 65 (20%), 52 (16%) and 18 (5·5%) patients, respectively. Regression analysis revealed frequent blood transfusions, indirect bilirubin and male gender as clinical predictors of SCN. APOL1 high-risk genotype (G1/G1, G2/G2 and G1/G2) was significantly associated with albuminuria (P = 0·04) and hyperfiltration (P = 0·001). HMOX1 GT-dinucleotide long repeats were significantly associated with lower eGFRcr. The study revealed a high burden of kidney damage among Congolese children and provided evidence of the possible role of APOL1 and HMOX1 in making children more susceptible to kidney complications.
Collapse
Affiliation(s)
- Oyindamola Christiana Adebayo
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium
| | - DieuMerci Kabasele Betukumesu
- Division of Nephrology, Department of Paediatrics, Faculty of Medicine, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Agathe Bikupe Nkoy
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium.,Division of Nephrology, Department of Paediatrics, Faculty of Medicine, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | | | - Pepe Mfutu Ekulu
- Division of Nephrology, Department of Paediatrics, Faculty of Medicine, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Lambertus P Van den Heuvel
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Paediatric Nephrology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Elena N Levtchenko
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Paediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Veerle Labarque
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Paediatric Haemato-Oncology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Detsika MG, Lianos EA. Hemopexin Modulates Expression of Complement Regulatory Proteins in Rat Glomeruli. Curr Issues Mol Biol 2021; 43:1081-1089. [PMID: 34563046 PMCID: PMC8928991 DOI: 10.3390/cimb43020077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 09/02/2021] [Indexed: 11/17/2022] Open
Abstract
In systemic hemolysis and in hematuric forms of kidney injury, the major heme scavenging protein, hemopexin (HPX), becomes depleted, and the glomerular microvasculature (glomeruli) is exposed to high concentrations of unbound heme, which, in addition to causing oxidative injury, can activate complement cascades; thus, compounding extent of injury. It is unknown whether unbound heme can also activate specific complement regulatory proteins that could defend against complement-dependent injury. Isolated rat glomeruli were incubated in media supplemented with HPX-deficient (HPX-) or HPX-containing (HPX+) sera as a means of achieving different degrees of heme partitioning between incubation media and glomerular cells. Expression of heme oxygenase (HO)-1 and of the complement activation inhibitors, decay-accelerating factor (DAF), CD59, and complement receptor-related gene Y (Crry), was assessed by western blot analysis. Expression of HO-1 and of the GPI-anchored DAF and CD59 proteins increased in isolated glomeruli incubated with HPX- sera with no effect on Crry expression. Exogenous heme (hemin) did not further induce DAF but increased Crry expression. HPX modulates heme-mediated induction of complement activation controllers in glomeruli. This effect could be of translational relevance in glomerular injury associated with hematuria.
Collapse
Affiliation(s)
- Maria G. Detsika
- 1st Department of Critical Care Medicine and Pulmonary Services, G. P. Livanou and M. Simou Laboratories, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10675 Athens, Greece
| | - Elias A. Lianos
- Veterans Affairs Medical Center and Virginia Tech., Carilion School of Medicine, 1970 Roanoke Blvd, Salem, VA 24153, USA;
| |
Collapse
|
28
|
Payán-Pernía S, Ruiz Llobet A, Remacha Sevilla ÁF, Egido J, Ballarín Castán JA, Moreno JA. Sickle cell nephropathy. Clinical manifestations and new mechanisms involved in kidney injury. Nefrologia 2021; 41:373-382. [PMID: 36165106 DOI: 10.1016/j.nefroe.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/03/2020] [Indexed: 06/16/2023] Open
Abstract
Kidney problems are among the most common complications in sickle cell disease (SCD). They occur early in childhood and are one of the main factors related to mortality in these patients. The main underlying pathogenic mechanisms are vaso-occlusion and haemolysis. The renal medulla has ideal conditions for the sickling of red cells due to its low partial pressure of oxygen, high osmolarity and acidic pH. Initially, sickle-cell formation in the vasa recta of the renal medulla causes hyposthenuria. This is universal and appears in early childhood. Microscopic and macroscopic haematuria also occur, in part related to renal papillary necrosis when the infarcts are extensive. Release of prostaglandins in the renal medulla due to ischaemia leads to an increase in the glomerular filtration rate (GFR). Adaptively, sodium reabsorption in the proximal tubule increases, accompanied by increased creatinine secretion. Therefore, the GFR estimated from creatinine may be overestimated. Focal segmental glomerulosclerosis is the most common glomerular disease. Albuminuria is very common and reduction has been found in 72.8% of subjects treated with ACE inhibitors or ARB. Recent evidence suggests that free haemoglobin has harmful effects on podocytes, and may be a mechanism involved in impaired kidney function in these patients. These effects need to be better studied in SCD, as they could provide a therapeutic alternative in sickle cell nephropathy.
Collapse
Affiliation(s)
- Salvador Payán-Pernía
- Unidad de Eritropatología, Unidad de Gestión Clínica de Hematología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC), Sevilla, Spain.
| | - Anna Ruiz Llobet
- Servicios y Unidades de Referencia (CSUR) de Eritropatología Hereditaria, Hospital Sant Joan de Déu - Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Servicio de Hematología Pediátrica, Hospital Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain; Institut de Recerca, Fundació Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Ángel Francisco Remacha Sevilla
- Servicios y Unidades de Referencia (CSUR) de Eritropatología Hereditaria, Hospital Sant Joan de Déu - Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Servicio de Hematología, Hospital de la Santa Creu i Sant Pau, Instituto de Investigación Biomédica Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jesús Egido
- Laboratorio de Enfermedades Renales, Vasculares y Diabetes, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
| | | | - Juan Antonio Moreno
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Instituto Maimónides de Investigaciones Biomédicas de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba, Spain.
| |
Collapse
|
29
|
Hsiao CC, Hou YS, Liu YH, Ko JY, Lee CT. Combined Melatonin and Extracorporeal Shock Wave Therapy Enhances Podocyte Protection and Ameliorates Kidney Function in a Diabetic Nephropathy Rat Model. Antioxidants (Basel) 2021; 10:antiox10050733. [PMID: 34066452 PMCID: PMC8148201 DOI: 10.3390/antiox10050733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 01/18/2023] Open
Abstract
(1) Background: Diabetic nephropathy (DN) is common complication of diabetes. Current therapy for DN does not include promotion of podocyte protection. Therefore, we investigated the therapeutic effect of melatonin (Mel) combined extracorporeal shock wave (SW) therapy on a DN rat model. (2) Methods: The DN rats were treated with Mel (5 mg/kg) twice a week for 6 weeks and SW treatment once a week (0.13 mJ/mm2) for 6 weeks. We assessed urine microalbumin, albumin to creatinine ratio (ACR), glomerular hypertrophy, glomerular fibrosis, podocyte markers (Wilm’s tumor protein-1, synaptopodin and nephrin), cell proliferation, cell survival, cell apoptosis, renal inflammation and renal oxidative stress. (3) Results: The Mel combined SW therapy regimen significantly reduced urine microalbumin excretion (3.3 ± 0.5 mg/dL, p < 0.001), ACR (65.2 ± 8.3 mg/g, p < 0.001), glomerular hypertrophy (3.1 ± 0.1 × 106 μm3, p < 0.01) and glomerular fibrosis (0.9 ± 0.4 relative mRNA fold, p < 0.05). Moreover, the Mel combined SW therapy regimen significantly increased podocyte number (44.1 ± 5.0% area of synaptopodin, p < 0.001) in the Mel combined SW group. This is likely primarily because Mel combined with SW therapy significantly reduced renal inflammation (753 ± 46 pg/mg, p < 0.01), renal oxidative stress (0.6 ± 0.04 relative density, p < 0.05), and apoptosis (0.3 ± 0.03 relative density, p < 0.001), and also significantly increased cell proliferation (2.0 ± 0.2% area proliferating cell nuclear antigen (PCNA), p < 0.01), cell survival, and nephrin level (4.2 ± 0.4 ng/mL, p < 0.001). (4) Conclusions: Mel combined SW therapy enhances podocyte protection and ameliorates kidney function in a DN rat model. Mel combined SW therapy may serve as a novel noninvasive and effective treatment of DN.
Collapse
Affiliation(s)
- Chang-Chun Hsiao
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-C.H.); (Y.-S.H.); (Y.-H.L.)
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - You-Syuan Hou
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-C.H.); (Y.-S.H.); (Y.-H.L.)
| | - Yu-Hsuan Liu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-C.H.); (Y.-S.H.); (Y.-H.L.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang-Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jih-Yang Ko
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang-Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Correspondence: ; Tel.: +886-7731-7123 (ext. 8306)
| |
Collapse
|
30
|
Taguchi S, Hidaka S, Yanai M, Ishioka K, Matsui K, Mochida Y, Moriya H, Ohtake T, Kobayashi S. Renal hemosiderosis presenting with acute kidney Injury and macroscopic hematuria in Immunoglobulin A nephropathy: a case report. BMC Nephrol 2021; 22:132. [PMID: 33858363 PMCID: PMC8048362 DOI: 10.1186/s12882-021-02334-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/02/2021] [Indexed: 11/19/2022] Open
Abstract
Background Macroscopic hematuria-associated acute kidney injury (AKI) is a well-known complication of immunoglobulin A (IgA) nephropathy. In such cases, intratubular obstruction by red blood cell (RBC) casts and acute tubular necrosis are mainly observed pathologically. Herein, we report the case of a patient with IgA nephropathy presenting with AKI following an episode of macrohematuria. The patient presented with severe renal tubular hemosiderosis and acute tubular necrosis and without any obvious obstructive RBC casts. Case presentation A 68-year-old woman, who was diagnosed with IgA nephropathy on renal biopsy 6 years ago, was admitted to our hospital after an episode of macroscopic glomerular hematuria and AKI following upper respiratory tract infection. Renal biopsy showed mesangial proliferation of the glomeruli, including crescent formation in 17 % of the glomeruli, and acute tubular necrosis without obvious hemorrhage or obstructive RBC casts. The application of Perls’ Prussian blue stain showed hemosiderin deposition in the renal proximal tubular cells. Immunofluorescence showed granular mesangial deposits of IgA and C3. Based on these findings, she was diagnosed with acute tubular necrosis with a concurrent IgA nephropathy flare-up. Moreover, direct tubular injury by heme and iron was considered to be the cause of AKI. She was treated with intravenous pulse methylprednisolone followed by oral prednisolone. Thereafter, the gross hematuria gradually faded, and her serum creatinine levels decreased. Conclusions IgA nephropathy presenting with acute kidney injury accompanied by macrohematuria may cause renal hemosiderosis and acute tubular necrosis without obstructive RBC casts. Hemosiderosis may be a useful indicator for determining the pathophysiology of macroscopic hematuria-associated AKI. However, renal hemosiderosis may remain undiagnosed. Thus, Perls’ Prussian blue iron staining should be more widely used in patients presenting with hematuria.
Collapse
Affiliation(s)
- Shinya Taguchi
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan. .,Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, 236-0004, Yokohama, Japan.
| | - Sumi Hidaka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan
| | - Mitsuru Yanai
- Department of Pathology, Sapporo Tokushukai Hospital, 1-1-1 Oyachi-higashi, Atsubetsu-ku, 004-0041, Sapporo, Hokkaido, Japan.,Hokkaido Renal Pathology Center, IT-FRONTBuilding, 28-196, N9W15, Chuo-ku, 060-0009, Sapporo, Hokkaido, Japan
| | - Kunihiro Ishioka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan
| | - Kenji Matsui
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan
| | - Yasuhiro Mochida
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan
| | - Hidekazu Moriya
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan
| | - Takayasu Ohtake
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan
| | - Shuzo Kobayashi
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, 1370-1 Okamoto, 247-8553, Kamakura, Kanagawa, Japan
| |
Collapse
|
31
|
Wlazlo E, Mehrad B, Morel L, Scindia Y. Iron Metabolism: An Under Investigated Driver of Renal Pathology in Lupus Nephritis. Front Med (Lausanne) 2021; 8:643686. [PMID: 33912577 PMCID: PMC8071941 DOI: 10.3389/fmed.2021.643686] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/10/2021] [Indexed: 12/13/2022] Open
Abstract
Nephritis is a common manifestation of systemic lupus erythematosus, a condition associated with inflammation and iron imbalance. Renal tubules are the work horse of the nephron. They contain a large number of mitochondria that require iron for oxidative phosphorylation, and a tight control of intracellular iron prevents excessive generation of reactive oxygen species. Iron supply to the kidney is dependent on systemic iron availability, which is regulated by the hepcidin-ferroportin axis. Most of the filtered plasma iron is reabsorbed in proximal tubules, a process that is controlled in part by iron regulatory proteins. This review summarizes tubulointerstitial injury in lupus nephritis and current understanding of how renal tubular cells regulate intracellular iron levels, highlighting the role of iron imbalance in the proximal tubules as a driver of tubulointerstitial injury in lupus nephritis. We propose a model based on the dynamic ability of iron to catalyze reactive oxygen species, which can lead to an accumulation of lipid hydroperoxides in proximal tubular epithelial cells. These iron-catalyzed oxidative species can also accentuate protein and autoantibody-induced inflammatory transcription factors leading to matrix, cytokine/chemokine production and immune cell infiltration. This could potentially explain the interplay between increased glomerular permeability and the ensuing tubular injury, tubulointerstitial inflammation and progression to renal failure in LN, and open new avenues of research to develop novel therapies targeting iron metabolism.
Collapse
Affiliation(s)
- Ewa Wlazlo
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, United States.,Department of Pathology, University of Florida, Gainesville, FL, United States
| | - Laurence Morel
- Department of Pathology, University of Florida, Gainesville, FL, United States
| | - Yogesh Scindia
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, United States.,Department of Pathology, University of Florida, Gainesville, FL, United States.,Division of Nephrology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
32
|
Abstract
Kidney problems are among the most common complications in sickle cell disease (SCD). They occur early in childhood and are one of the main factors related to mortality in these patients. The main underlying pathogenic mechanisms are vaso-occlusion and haemolysis. The renal medulla has ideal conditions for the sickling of red cells due to its low partial pressure of oxygen, high osmolarity and acidic pH. Initially, sickle-cell formation in the vasa recta of the renal medulla causes hyposthenuria. This is universal and appears in early childhood. Microscopic and macroscopic haematuria also occur, in part related to renal papillary necrosis when the infarcts are extensive. Release of prostaglandins in the renal medulla due to ischaemia leads to an increase in the glomerular filtration rate (GFR). Adaptively, sodium reabsorption in the proximal tubule increases, accompanied by increased creatinine secretion. Therefore, the GFR estimated from creatinine may be overestimated. Focal segmental glomerulosclerosis is the most common glomerular disease. Albuminuria is very common and reduction has been found in 72.8% of subjects treated with ACE inhibitors or ARB. Recent evidence suggests that free haemoglobin has harmful effects on podocytes, and may be a mechanism involved in impaired kidney function in these patients. These effects need to be better studied in SCD, as they could provide a therapeutic alternative in sickle cell nephropathy.
Collapse
|
33
|
Pesce F, Stea ED, Rossini M, Fiorentino M, Piancone F, Infante B, Stallone G, Castellano G, Gesualdo L. Glomerulonephritis in AKI: From Pathogenesis to Therapeutic Intervention. Front Med (Lausanne) 2021; 7:582272. [PMID: 33738291 PMCID: PMC7960664 DOI: 10.3389/fmed.2020.582272] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is increasingly emerging as a global emergency. Sepsis, major surgery, and nephrotoxic drugs are the main causes of AKI in hospitalized patients. However, glomerulonephritis accounts for about 10% of AKI episodes in adults, mainly related to rapidly progressive glomerulonephritis resulting from granulomatous polyangiitis (GPA, Wegener granulomatosis), microscopic polyangiitis (MPA), and anti-glomerular basement membrane (GBM) disease. Also, diffuse proliferative lupus nephritis, immunoglobulin A nephropathy, post-streptococcal glomerulonephritis, mixed cryoglobulinemia, mesangiocapillary glomerulonephritis, membranous nephropathy, hemolytic uremic syndrome, thrombotic thrombocytopenic purpura, and scleroderma can induce acute renal failure. Early diagnosis of AKI due to glomerulonephritis is crucial for prompt, effective management to improve short- and long-term outcomes. Kidney biopsy is the gold standard for the diagnosis of glomerular disease, but it is not frequently performed in critically ill patients because of their clinical conditions. In this setting, a growing number of diagnostic assays can support the working hypothesis, including antineutrophil cytoplasmic antibodies (ANCAs), anti-double-stranded DNA antibodies, anti-GBM antibodies, antistreptolysin O and anti-DNase B antibodies, cryoglobulins, antiphospholipid antibodies, and complement levels. Therapeutic strategies in AKI patients with glomerulonephritis include high-dose corticosteroids, cyclophosphamide, and plasma exchange. This article reviews the wide spectrum of glomerulopathies associated with AKI, describing the immunological mechanisms underlying glomerular diseases and presenting an overview of the therapeutic options.
Collapse
Affiliation(s)
- Francesco Pesce
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Emma D Stea
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Michele Rossini
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Marco Fiorentino
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Fausta Piancone
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Science, University of Foggia, Foggia, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Science, University of Foggia, Foggia, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Science, University of Foggia, Foggia, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| |
Collapse
|
34
|
Gomez-Fernández P, Martín Santana A, Arjona Barrionuevo JDD. Oral anticoagulation in chronic kidney disease with atrial fibrillation. Nefrologia 2021; 41:137-153. [PMID: 36165375 DOI: 10.1016/j.nefroe.2021.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/11/2020] [Indexed: 06/16/2023] Open
Abstract
Chronic kidney disease (CKD) and atrial fibrillation (AF) frequently coexist, amplifying the risk of cardiovascular events and mortality. In patients with CKD stage 3 and non-valvular AF, direct oral anticoagulants (DOACs) have shown, compared to vitamin K antagonists (VKA), equal or greater efficacy in the prevention of stroke and systemic embolism, and greater safety. There are no randomizedtrials of the efficacy and safety of DOACs and VKA in advanced CKD. On the other hand, observational studies suggest that DOACs, compared to warfarin, are associated with a lower risk of acute kidney damage and generation/progression of CKD. This paper reviews the epidemiological and pathophysiological aspects of the CKD and AF association, the evidence of the efficacy and safety of warfarin and ACODs in various stages of CKD with AF as well as the comparison between warfarin and ACODs in efficacy and anticoagulant safety, and in its renal effects.
Collapse
Affiliation(s)
- Pablo Gomez-Fernández
- Unidad de Factores de Riesgo Vascular, Servicio de Nefrología, Hospital Universitario de Jerez, Jerez de la Frontera, Cádiz, Spain.
| | | | | |
Collapse
|
35
|
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: 21] [Impact Index Per Article: 5.3] [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
|
36
|
Guerrero-Hue M, Rayego-Mateos S, Vázquez-Carballo C, Palomino-Antolín A, García-Caballero C, Opazo-Rios L, Morgado-Pascual JL, Herencia C, Mas S, Ortiz A, Rubio-Navarro A, Egea J, Villalba JM, Egido J, Moreno JA. Protective Role of Nrf2 in Renal Disease. Antioxidants (Basel) 2020; 10:antiox10010039. [PMID: 33396350 PMCID: PMC7824104 DOI: 10.3390/antiox10010039] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the fastest-growing causes of death and is predicted to become by 2040 the fifth global cause of death. CKD is characterized by increased oxidative stress and chronic inflammation. However, therapies to slow or prevent CKD progression remain an unmet need. Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor that plays a key role in protection against oxidative stress and regulation of the inflammatory response. Consequently, the use of compounds targeting Nrf2 has generated growing interest for nephrologists. Pre-clinical and clinical studies have demonstrated that Nrf2-inducing strategies prevent CKD progression and protect from acute kidney injury (AKI). In this article, we review current knowledge on the protective mechanisms mediated by Nrf2 against kidney injury, novel therapeutic strategies to induce Nrf2 activation, and the status of ongoing clinical trials targeting Nrf2 in renal diseases.
Collapse
Affiliation(s)
- Melania Guerrero-Hue
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Sandra Rayego-Mateos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Cristina Vázquez-Carballo
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
| | - Alejandra Palomino-Antolín
- Research Unit, Hospital Universitario Santa Cristina, IIS-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (A.P.-A.); (J.E.)
- Departament of Pharmacology and Therapeutics, Medicine Faculty, Instituto Teófilo Hernando, Autónoma University, 28029 Madrid, Spain
| | - Cristina García-Caballero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Lucas Opazo-Rios
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - José Luis Morgado-Pascual
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
| | - Carmen Herencia
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
| | - Sebastián Mas
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Red Nacional Investigaciones Nefrológicas (REDINREN), 28040 Madrid, Spain
| | - Alfonso Rubio-Navarro
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Javier Egea
- Research Unit, Hospital Universitario Santa Cristina, IIS-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (A.P.-A.); (J.E.)
- Departament of Pharmacology and Therapeutics, Medicine Faculty, Instituto Teófilo Hernando, Autónoma University, 28029 Madrid, Spain
| | - José Manuel Villalba
- Department of Cell Biology, Physiology, and Immunology, Agrifood Campus of International Excellence (ceiA3), University of Cordoba, 14014 Cordoba, Spain;
| | - Jesús Egido
- Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Autónoma University, 28040 Madrid, Spain; (C.V.-C.); (L.O.-R.); (C.H.); (S.M.); (A.O.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain; (M.G.-H.); (S.R.-M.); (C.G.-C.); (J.L.M.-P.)
- Department of Cell Biology, Physiology, and Immunology, Agrifood Campus of International Excellence (ceiA3), University of Cordoba, 14014 Cordoba, Spain;
- Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
- Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-957-218-039
| |
Collapse
|
37
|
Trimarchi H, Ortiz A, Sánchez-Niño MD. Lyso-Gb3 Increases αvβ3 Integrin Gene Expression in Cultured Human Podocytes in Fabry Nephropathy. J Clin Med 2020; 9:jcm9113659. [PMID: 33203029 PMCID: PMC7696179 DOI: 10.3390/jcm9113659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Podocyturia in Fabry nephropathy leads to glomerulosclerosis and kidney disease progression. Integrins are involved in podocyte attachment to the glomerular basement membrane. We hypothesized that in Fabry nephropathy, lyso-Gb3 could modulate αvβ3 expression in podocytes. Together with UPAR, the αvβ3 integrin is a key mechanism involved in podocyte detachment and podocyturia. Methods: In cultured human podocytes stimulated with lyso-Gb3, the mRNA expression of the ITGAV and ITGB3 genes encoding integrins αv and β3, respectively, was analyzed by RT-qPCR. Results: In cultured human podocytes, lyso-Gb3 at concentrations encountered in the serum of Fabry patients increased ITGAV and ITGB3 mRNA levels within 3 to 6 h. This pattern of gene expression is similar to that previously observed for PLAUR (UPAR) gene expression but is in contrast to the delayed (24 h) upregulation of other markers of podocyte stress and mediators of injury, such as CD80, TGFβ1, CD74, Notch1, and HES. Conclusions: Human podocyte stress in response to glycolipid overload in Fabry nephropathy, exemplified by lyso-Gb3, is characterized by an early increase in the expression of components of the αvβ3/UPAR system, which contrasts with the delayed rise in the expression of other mediators of podocyte injury. This suggests that the αvβ3/UPAR system may be a therapeutic target in Fabry nephropathy.
Collapse
Affiliation(s)
- Hernán Trimarchi
- Nephrology Service, Hospital Británico de Buenos Aires, 1280 Buenos Aires, Argentina
- Correspondence: ; Tel.: +1280-5411-4309-6400
| | - Alberto Ortiz
- IIS-Fundación Jimenez Díaz, School of Medicine, UAM, 28040 Madrid, Spain; (A.O.); (M.D.S.-N.)
- Spanish Renal Research Network (REDINREN), 28004 Madrid, Spain
| | - Maria Dolores Sánchez-Niño
- IIS-Fundación Jimenez Díaz, School of Medicine, UAM, 28040 Madrid, Spain; (A.O.); (M.D.S.-N.)
- Spanish Renal Research Network (REDINREN), 28004 Madrid, Spain
- Pharmacology and Therapeutics Department, Universidad Autonoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
38
|
Wei W, Ma N, Fan X, Yu Q, Ci X. The role of Nrf2 in acute kidney injury: Novel molecular mechanisms and therapeutic approaches. Free Radic Biol Med 2020; 158:1-12. [PMID: 32663513 DOI: 10.1016/j.freeradbiomed.2020.06.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/24/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
Acute kidney injury (AKI) is a common clinical syndrome that is related to high morbidity and mortality. Oxidative stress, including the production of reactive oxygen species (ROS), appears to be the main element in the occurrence of AKI and the cause of the progression of chronic kidney disease (CKD) into end-stage renal disease (ESRD). Nuclear factor erythroid 2 related factor 2 (Nrf2) is a significant regulator of redox balance that has been shown to improve kidney disease by eliminating ROS. To date, researchers have found that the use of Nrf2-activated compounds can effectively reduce ROS, thereby preventing or retarding the progression of various types of AKI. In this review, we summarized the molecular mechanisms of Nrf2 and ROS in AKI and described the latest findings on the therapeutic potential of Nrf2 activators in various types of AKI.
Collapse
Affiliation(s)
- Wei Wei
- Department of Urology, The First Hospital, Jilin University, Changchun, China
| | - Ning Ma
- Department of Urology, The First Hospital, Jilin University, Changchun, China
| | - Xiaoye Fan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Qinlei Yu
- Jilin Provincial Animal Disease Control Center, 4510 Xi'an Road, Changchun, 130062, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
39
|
Gutiérrez E, Carvaca-Fontán F, Luzardo L, Morales E, Alonso M, Praga M. A Personalized Update on IgA Nephropathy: A New Vision and New Future Challenges. Nephron Clin Pract 2020; 144:555-571. [PMID: 32818944 DOI: 10.1159/000509997] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 07/08/2020] [Indexed: 11/19/2022] Open
Abstract
IgA nephropathy (IgAN) is the most common primary glomerulonephritis in the world among patients undergoing renal biopsy. Approximately 30% of patients with IgAN develop end-stage kidney disease 20 years after renal biopsy. It is a glomerulopathy with a very broad clinical presentation, making it difficult to stratify and treat. IgAN is characterized by dysregulation of the immune system, which causes an abnormal synthesis of IgA1 that is deglycosylated causing its mesangial deposition. IgAN pathogenesis is incompletely understood; the current multi-hit hypothesis of IgAN pathogenesis does not explain the range of glomerular inflammation and renal injury associated with mesangial IgA deposition. Although associations between IgAN and glomerular and circulating markers of complement activation are established, the mechanism of complement activation and contribution to glomerular inflammation and injury are not defined. On the other hand, the renal-gut connection can also play an important role in the pathogenesis of IgAN with possible therapeutic implications. In order to standardize the histological findings, the Oxford Classification has allowed clarifying renal lesions that confer potential risk of progression. Currently, except for the blockade of the renin-angiotensin-aldosterone system, no other therapies are available in clinical setting for the treatment of IgAN, although the range of new drugs under investigation is extensive. The incorporation in the next trials of clinical parameters such as the amount of hematuria and histological lesions may allow more personalized therapeutic approaches. To summarize, in recent years, several important efforts have taken place in the understanding of IgAN, but still, further studies are warranted to elucidate the best therapeutic strategies according to the risk to improve the prognosis of this entity.
Collapse
Affiliation(s)
- Eduardo Gutiérrez
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain, .,Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain,
| | - Fernando Carvaca-Fontán
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Leonella Luzardo
- Department of Nephrology and Pathophysiology, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Enrique Morales
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Marina Alonso
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,Department of Pathological Anatomy, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Manuel Praga
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
40
|
Gembillo G, Siligato R, Cernaro V, Santoro D. Complement Inhibition Therapy and Dialytic Strategies in Paroxysmal Nocturnal Hemoglobinuria: The Nephrologist's Opinion. J Clin Med 2020; 9:1261. [PMID: 32357555 PMCID: PMC7287718 DOI: 10.3390/jcm9051261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/24/2022] Open
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare clonal disease that presents an estimated incidence of 1.3 cases per million per year, with a prevalence of 15.9 cases per million. It is characterized by hemolysis, bone marrow dysfunction with peripheral blood cytopenia, hypercoagulability, thrombosis, renal impairment and arterial and pulmonary hypertension. Hemolysis and subsequent hemosiderin accumulation in tubular epithelium cells induce tubular atrophy and interstitial fibrosis. The origin of PNH is the somatic mutation in the X-linked phosphatidylinositol glycan class A (PIG-A) gene located on Xp22: this condition leads to the production of clonal blood cells with a deficiency in those surface proteins that protect against the lytic action of the activated complement system. Despite the increased knowledge of this syndrome, therapies for PNH were still only experimental and symptomatic, until the introduction of the C5 complement blockade agent Eculizumab. A second generation of anti-complement agents is currently under investigation, representing future promising therapeutic strategies for patients affected by PNH. In the case of chronic hemolysis and renal iron deposition, a multidisciplinary approach should be considered to avoid or treat acute tubular injury or acute kidney injury (AKI). New promising perspectives derive from complement inhibitors and iron chelators, as well as more invasive treatments such as immunoadsorption or the use of dedicated hemodialysis filters in the presence of AKI.
Collapse
Affiliation(s)
- Guido Gembillo
- Unit of Nephrology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (R.S.); (V.C.); (D.S.)
| | | | | | | |
Collapse
|
41
|
Poulaki E, Detsika MG, Fourtziala E, Lianos EA, Gakiopoulou H. Podocyte-targeted Heme Oxygenase (HO)-1 overexpression exacerbates age-related pathology in the rat kidney. Sci Rep 2020; 10:5719. [PMID: 32235880 PMCID: PMC7109035 DOI: 10.1038/s41598-020-62016-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/20/2020] [Indexed: 11/09/2022] Open
Abstract
Although Heme Oxygenase-1 (HO-1) induction in various forms of kidney injury is protective, its role in age-related renal pathology is unknown. In the ageing kidney there is nephron loss and lesions of focal glomerulosclerosis, interstitial fibrosis, tubular atrophy and arteriolosclerosis. Underlying mechanisms include podocyte (visceral glomerular epithelial cell/GEC) injury. To assess whether HO-1 can attenuate ageing - related lesions, rats with GEC-targeted HO-1 overexpression (GECHO-1 rats) were generated using a Sleeping Beauty (SB) transposon system and extent of lesions over a 12-month period were assessed and compared to those in age-matched wild-type (WT) controls. GECHO-1 rats older than 6 months developed albuminuria that was detectable at 6 months and became significantly higher compared to age-matched WT controls at 12 months. In GECHO-1 rats, lesions of focal segmental and global glomerulosclerosis as well as tubulointerstitial lesions were prominent while podocytes were edematous with areas of foot process effacement and glomerular basement membrane thickening and wrinkling. GECHO-1 rats also developed hemoglobinuria and hemosiderinuria associated with marked tubular hemosiderin deposition and HO-1 induction, while there was depletion of splenic iron stores. Kidney injury was of sufficient magnitude to increase serum lactate dehydrogenase (LDH) and was oxidative in nature as shown by increased expression of 8-hydroxydeoxyguanosine (8-OHdg, a byproduct of oxidative DNA damage) in podocytes and tubular epithelial cells. These observations highlight a detrimental effect of podocyte-targeted HO-1 overexpression on ageing-related renal pathology and point to increased renal iron deposition as a putative underlying mechanism.
Collapse
Affiliation(s)
- Elpida Poulaki
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens, 11527, Greece
| | - Maria G Detsika
- First Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, National and Kapodistrian University School of Medicine, 3 Ploutarchou Street, Athens, 10675, Greece
| | - Eythimia Fourtziala
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens, 11527, Greece
| | - Elias A Lianos
- Veterans Affairs Medical Center and Virginia Tech. Carilion School of Medicine, 1970 Roanoke Blvd, Salem, VA, 24153, USA.
| | - Hariklia Gakiopoulou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens, 11527, Greece
| |
Collapse
|
42
|
The effects of Hemiscorpius lepturus induced-acute kidney injury on PGC-1α gene expression: From induction to suppression in mice. Toxicon 2019; 174:57-63. [PMID: 31887316 DOI: 10.1016/j.toxicon.2019.12.154] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/05/2019] [Accepted: 12/21/2019] [Indexed: 11/23/2022]
Abstract
Hemiscorpius lepturus envenomation induces acute kidney injury (AKI) through hemoglubinoria and mitochondrial dysfunction. Mitochondria supports ATP production to promote the regulation of fluid and electrolyte balance. Mitochondrial homeostasis in different metabolic environments can be adjusted by overexpression of PGC-1α. High reactive oxygen species (ROS) production after H. lepturus envenomation and heme oxygenase-1 (HO-1) overexpression causes ATP depletion as well as mitochondrial homeostasis disruption, which lead to progression in renal diseases. The present study aims to evaluate the role of venom induced-AKI in modulating mitochondrial function in cell death and metabolic signaling associated with PPAR-α, PGC-1α, and Nrf-2 as the main transcription factors involved in metabolism. Based on the data, two significant events occurred after envenomation: reduction of gl glutathione level and overexpression of the cytoprotective enzyme HO-1. Apaoptosis induction is associated with a significant decrease in the transcription of PPAR-α, PGC-1α and Nrf-2 after administrating lethal dose of venom (10 mg/kg). Furthermore, at the lower doses of venom (1 and 5 mg/kg), with a significant recovery accompanied with PGC-1α upregulation occurs after AKI. As the findings indicate, PGC-1α has a key role in restoring the mitochondrial function at the recovery phase of mouse model of AKI, which highlights the PGC-1α as a therapeutic target for venom induced-AKI prevention and treatment.
Collapse
|
43
|
|
44
|
Abstract
Iron is an essential element that is indispensable for life. The delicate physiological body iron balance is maintained by both systemic and cellular regulatory mechanisms. The iron-regulatory hormone hepcidin assures maintenance of adequate systemic iron levels and is regulated by circulating and stored iron levels, inflammation and erythropoiesis. The kidney has an important role in preventing iron loss from the body by means of reabsorption. Cellular iron levels are dependent on iron import, storage, utilization and export, which are mainly regulated by the iron response element-iron regulatory protein (IRE-IRP) system. In the kidney, iron transport mechanisms independent of the IRE-IRP system have been identified, suggesting additional mechanisms for iron handling in this organ. Yet, knowledge gaps on renal iron handling remain in terms of redundancy in transport mechanisms, the roles of the different tubular segments and related regulatory processes. Disturbances in cellular and systemic iron balance are recognized as causes and consequences of kidney injury. Consequently, iron metabolism has become a focus for novel therapeutic interventions for acute kidney injury and chronic kidney disease, which has fuelled interest in the molecular mechanisms of renal iron handling and renal injury, as well as the complex dynamics between systemic and local cellular iron regulation.
Collapse
|
45
|
Esteras R, Cannata-Ortiz P, Del Palacio-Tamarit M, Guerrero-Hue M, García-Caballero C, Egido J, Gimeno J, Ortiz A, Gracia-Iguacel C, Moreno JA. Podocyte and tubular involvement in AngioJet-induced kidney injury. Clin Kidney J 2019; 14:424-428. [PMID: 33564448 PMCID: PMC7857840 DOI: 10.1093/ckj/sfz104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 07/10/2019] [Indexed: 11/14/2022] Open
Abstract
The AngioJet technique combines localized thrombolysis and percutaneous mechanical thrombectomy (PMT). However, PMT may cause acute kidney injury (AKI), which has been ascribed to severe mechanical haemolysis, although no renal biopsies have been reported. We now report the first renal biopsy in a patient with AKI following PMT. There is histological evidence of haemoglobin (Hb)-induced tubular injury and podocyte stress characterized by intracellular Hb and staining for ferritin and hemo-oxygenase-1, suggestive of an adaptive response to oxidative stress. This confirms that Hb is involved in kidney cell injury and supports the existence of several different kidney cellular targets.
Collapse
Affiliation(s)
- Raquel Esteras
- Department of Nephrology, Fundacion Jimenez Diaz Health Research Institute (IIS-FJD), Autonoma University of Madrid, Madrid, Spain.,Red de Investigacion Renal, Madrid, Spain
| | - Pablo Cannata-Ortiz
- Red de Investigacion Renal, Madrid, Spain.,Department of Pathology, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
| | | | - Melania Guerrero-Hue
- Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz Health Research Institute, Autonoma University of Madrid, Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
| | - Cristina García-Caballero
- Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz Health Research Institute, Autonoma University of Madrid, Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
| | - Jesús Egido
- Department of Nephrology, Fundacion Jimenez Diaz Health Research Institute (IIS-FJD), Autonoma University of Madrid, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Javier Gimeno
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Alberto Ortiz
- Department of Nephrology, Fundacion Jimenez Diaz Health Research Institute (IIS-FJD), Autonoma University of Madrid, Madrid, Spain.,Red de Investigacion Renal, Madrid, Spain
| | - Carolina Gracia-Iguacel
- Department of Nephrology, Fundacion Jimenez Diaz Health Research Institute (IIS-FJD), Autonoma University of Madrid, Madrid, Spain.,Red de Investigacion Renal, Madrid, Spain
| | - Juan Antonio Moreno
- Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz Health Research Institute, Autonoma University of Madrid, Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Maimonides Biomedical Research Institute of Cordoba, University of Córdoba, Córdoba, Spain
| |
Collapse
|
46
|
Sevillano AM, Diaz M, Caravaca-Fontán F, Barrios C, Bernis C, Cabrera J, Calviño J, Castillo L, Cobelo C, Delgado-Mallén P, Espinosa M, Fernandez-Juarez G, Fernandez-Reyes MJ, Garcia-Osuna R, Garcia P, Goicoechea M, Gonzalez-Cabrera F, Guzmán DA, Heras M, Martín-Reyes G, Martinez A, Olea T, Peña JK, Quintana LF, Rabasco C, López Revuelta K, Rodas L, Rodriguez-Mendiola N, Rodriguez E, San Miguel L, Sanchez de la Nieta MD, Shabaka A, Sierra M, Valera A, Velo M, Verde E, Ballarin J, Noboa O, Moreno JA, Gutiérrez E, Praga M, on behalf of the Spanish Group for the Study of Glomerular Diseases (GLOSEN). IgA Nephropathy in Elderly Patients. Clin J Am Soc Nephrol 2019; 14:1183-1192. [PMID: 31311818 PMCID: PMC6682823 DOI: 10.2215/cjn.13251118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 05/01/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVES Some studies suggest that the incidence of IgA nephropathy is increasing in older adults, but there is a lack of information about the epidemiology and behavior of the disease in that age group. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In this retrospective multicentric study, we analyzed the incidence, forms of presentation, clinical and histologic characteristics, treatments received, and outcomes in a cohort of 151 patients ≥65 years old with biopsy-proven IgA nephropathy diagnosed between 1990 and 2015. The main outcome was a composite end point of kidney replacement therapy or death before kidney replacement therapy. RESULTS We found a significant increase in the diagnosis of IgA nephropathy over time from six patients in 1990-1995 to 62 in 2011-2015 (P value for trend =0.03). After asymptomatic urinary abnormalities (84 patients; 55%), AKI was the most common form of presentation (61 patients; 40%). Within the latter, 53 (86%) patients presented with hematuria-related AKI (gross hematuria and tubular necrosis associated with erythrocyte casts as the most important lesions in kidney biopsy), and eight patients presented with crescentic IgA nephropathy. Six (4%) patients presented with nephrotic syndrome. Among hematuria-related AKI, 18 (34%) patients were receiving oral anticoagulants, and this proportion rose to 42% among the 34 patients older than 72 years old who presented with hematuria-related AKI. For the whole cohort, survival rates without the composite end point were 74%, 48%, and 26% at 1, 2, and 5 years, respectively. Age, serum creatinine at presentation, and the degree of interstitial fibrosis in kidney biopsy were risk factors significantly associated with the outcome, whereas treatment with renin-angiotensin-aldosterone blockers was associated with a lower risk. Immunosuppressive treatments were not significantly associated with the outcome. CONCLUSIONS The diagnosis of IgA nephropathy among older adults in Spain has progressively increased in recent years, and anticoagulant therapy may be partially responsible for this trend. Prognosis was poor. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2019_07_16_CJASNPodcast_19_08_.mp3.
Collapse
Affiliation(s)
- Angel M. Sevillano
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| | - Monserrat Diaz
- Department of Nephrology, Fundación Puigvert, Barcelona, Spain
| | - Fernando Caravaca-Fontán
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| | - Clara Barrios
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Carmen Bernis
- Department of Nephrology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Jimena Cabrera
- Programa de prevención y tratamiento de las glomerulopatias (PPTG) de Uruguay, Sociedad Uruguaya de Nefrologia Centro de Nefrología, Hospital de Clinicas Montevideo, Montevideo, Uruguay
| | - Jesus Calviño
- Department of Nephrology, Hospital de Lucus Augusti, Lugo, Spain
| | - Lorena Castillo
- Department of Nephrology, Hospital Universitario Joan XXIII, Tarragona, Spain
| | - Carmen Cobelo
- Department of Nephrology, Hospital de Lucus Augusti, Lugo, Spain
| | | | - Mario Espinosa
- Department of Nephrology, Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Gema Fernandez-Juarez
- Department of Nephrology, Hospital Universitario Fundación Alcorcón, Alcorcon, Spain
| | | | | | - Patricia Garcia
- Department of Nephrology, Hospital Virgen de la Victoria, Malaga, Spain
| | - Marian Goicoechea
- Department of Nephrology, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Fayna Gonzalez-Cabrera
- Department of Nephrology, Hospital Universitario de Gran Canaria Dr. Negrin, Gran Canaria, Spain
| | - Diomaris A. Guzmán
- Department of Nephrology, Hospital Regional Universitario de Málaga, Malaga, Spain
| | - Manuel Heras
- Department of Nephrology, Hospital General de Segovia, Segovia, Spain
| | | | - Alberto Martinez
- Department of Nephrology, Hospital Universitario Joan XXIII, Tarragona, Spain
| | - Teresa Olea
- Department of Nephrology, Hospital Universitario La Paz, Madrid, Spain
| | - Jessy Korina Peña
- Department of Nephrology, Hospital Principe de Asturias, Alcala de Henares, Spain
| | - Luis F. Quintana
- Department of Nephrology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Cristina Rabasco
- Department of Nephrology, Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Katia López Revuelta
- Department of Nephrology, Hospital Universitario Fundación Alcorcón, Alcorcon, Spain
| | - Lida Rodas
- Department of Nephrology, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Eva Rodriguez
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Luz San Miguel
- Department of Nephrology, Fundación Puigvert, Barcelona, Spain
| | | | - Amir Shabaka
- Department of Nephrology, Hospital Clínico de Madrid, Madrid, Spain
| | - Milagros Sierra
- Department of Nephrology, Hospital San Pedro, Logrono, Spain
| | - Alfonso Valera
- Department of Nephrology, Hospital Virgen de la Victoria, Malaga, Spain
| | - Mercedes Velo
- Department of Nephrology, Hospital Clínico de Madrid, Madrid, Spain
| | - Eduardo Verde
- Department of Nephrology, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Jose Ballarin
- Department of Nephrology, Fundación Puigvert, Barcelona, Spain
| | - Oscar Noboa
- Nephrology Center, Hospital de Clínicas, Department of Medicine, Republic University, Montevideo, Uruguay; and
| | - Juan Antonio Moreno
- Department of Cell Biology, Physiology, and Immunology, Maimonides Biomedical Research Institute of Cordoba, University of Cordoba, Cordoba, Spain
| | - Eduardo Gutiérrez
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| | - on behalf of the Spanish Group for the Study of Glomerular Diseases (GLOSEN)
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
- Department of Nephrology, Fundación Puigvert, Barcelona, Spain
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Department of Nephrology, Hospital Universitario de La Princesa, Madrid, Spain
- Programa de prevención y tratamiento de las glomerulopatias (PPTG) de Uruguay, Sociedad Uruguaya de Nefrologia Centro de Nefrología, Hospital de Clinicas Montevideo, Montevideo, Uruguay
- Department of Nephrology, Hospital de Lucus Augusti, Lugo, Spain
- Department of Nephrology, Hospital Universitario Joan XXIII, Tarragona, Spain
- Department of Nephrology, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
- Department of Nephrology, Hospital Universitario Reina Sofia, Cordoba, Spain
- Department of Nephrology, Hospital Universitario Fundación Alcorcón, Alcorcon, Spain
- Department of Nephrology, Hospital General de Segovia, Segovia, Spain
- Department of Nephrology, Hospital de Palamós, Palamos, Spain
- Department of Nephrology, Hospital Virgen de la Victoria, Malaga, Spain
- Department of Nephrology, Hospital Universitario Gregorio Marañón, Madrid, Spain
- Department of Nephrology, Hospital Universitario de Gran Canaria Dr. Negrin, Gran Canaria, Spain
- Department of Nephrology, Hospital Regional Universitario de Málaga, Malaga, Spain
- Department of Nephrology, Hospital Universitario La Paz, Madrid, Spain
- Department of Nephrology, Hospital Principe de Asturias, Alcala de Henares, Spain
- Department of Nephrology, Hospital Clinic de Barcelona, Barcelona, Spain
- Department of Nephrology, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Department of Nephrology, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
- Department of Nephrology, Hospital Clínico de Madrid, Madrid, Spain
- Department of Nephrology, Hospital San Pedro, Logrono, Spain
- Nephrology Center, Hospital de Clínicas, Department of Medicine, Republic University, Montevideo, Uruguay; and
- Department of Cell Biology, Physiology, and Immunology, Maimonides Biomedical Research Institute of Cordoba, University of Cordoba, Cordoba, Spain
| |
Collapse
|
47
|
Rubio-Navarro A, Vázquez-Carballo C, Guerrero-Hue M, García-Caballero C, Herencia C, Gutiérrez E, Yuste C, Sevillano Á, Praga M, Egea J, Cannata P, Cortegano I, de Andrés B, Gaspar ML, Cadenas S, Michalska P, León R, Ortiz A, Egido J, Moreno JA. Nrf2 Plays a Protective Role Against Intravascular Hemolysis-Mediated Acute Kidney Injury. Front Pharmacol 2019; 10:740. [PMID: 31333462 PMCID: PMC6619398 DOI: 10.3389/fphar.2019.00740] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Massive intravascular hemolysis is associated with acute kidney injury (AKI). Nuclear factor erythroid-2-related factor 2 (Nrf2) plays a central role in the defense against oxidative stress by activating the expression of antioxidant proteins. We investigated the role of Nrf2 in intravascular hemolysis and whether Nrf2 activation protected against hemoglobin (Hb)/heme-mediated renal damage in vivo and in vitro. We observed renal Nrf2 activation in human hemolysis and in an experimental model of intravascular hemolysis promoted by phenylhydrazine intraperitoneal injection. In wild-type mice, Hb/heme released from intravascular hemolysis promoted AKI, resulting in decreased renal function, enhanced expression of tubular injury markers (KIM-1 and NGAL), oxidative and endoplasmic reticulum stress (ER), and cell death. These features were more severe in Nrf2-deficient mice, which showed decreased expression of Nrf2-related antioxidant enzymes, including heme oxygenase 1 (HO-1) and ferritin. Nrf2 activation with sulforaphane protected against Hb toxicity in mice and cultured tubular epithelial cells, ameliorating renal function and kidney injury and reducing cell stress and death. Nrf2 genotype or sulforaphane treatment did not influence the severity of hemolysis. In conclusion, our study identifies Nrf2 as a key molecule involved in protection against renal damage associated with hemolysis and opens novel therapeutic approaches to prevent renal damage in patients with severe hemolytic crisis. These findings provide new insights into novel aspects of Hb-mediated renal toxicity and may have important therapeutic implications for intravascular hemolysis-related diseases.
Collapse
Affiliation(s)
- Alfonso Rubio-Navarro
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Cristina Vázquez-Carballo
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Melania Guerrero-Hue
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Cristina García-Caballero
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Carmen Herencia
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | | | - Claudia Yuste
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| | - Ángel Sevillano
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| | - Javier Egea
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain.,Hospital Santa Cristina, Madrid, Spain
| | - Pablo Cannata
- Pathology Department, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Isabel Cortegano
- Immunology Department, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Belén de Andrés
- Immunology Department, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Luisa Gaspar
- Immunology Department, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Susana Cadenas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Patrycja Michalska
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Rafael León
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Alberto Ortiz
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Juan Antonio Moreno
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, Cordoba, Spain
| |
Collapse
|
48
|
Glomerular Hematuria: Cause or Consequence of Renal Inflammation? Int J Mol Sci 2019; 20:ijms20092205. [PMID: 31060307 PMCID: PMC6539976 DOI: 10.3390/ijms20092205] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/21/2019] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
Glomerular hematuria is a cardinal symptom of renal disease. Glomerular hematuria may be classified as microhematuria or macrohematuria according to the number of red blood cells in urine. Recent evidence suggests a pathological role of persistent glomerular microhematuria in the progression of renal disease. Moreover, gross hematuria, or macrohematuria, promotes acute kidney injury (AKI), with subsequent impairment of renal function in a high proportion of patients. In this pathological context, hemoglobin, heme, or iron released from red blood cells in the urinary space may cause direct tubular cell injury, oxidative stress, pro-inflammatory cytokine production, and further monocyte/macrophage recruitment. The aim of this manuscript is to review the role of glomerular hematuria in kidney injury, the role of inflammation as cause and consequence of glomerular hematuria, and to discuss novel therapies to combat hematuria.
Collapse
|
49
|
Jiao H, Xie D, Qiao Y. LncRNA PRINS is involved in the development of nephropathy in patients with diabetes via interaction with Smad7. Exp Ther Med 2019; 17:3203-3208. [PMID: 30936994 PMCID: PMC6434383 DOI: 10.3892/etm.2019.7307] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNA psoriasis-susceptibilityrelated RNA gene induced by stress (PRINS) is known to be involved in kidney ischemia reperfusion injury. The aim of the current study was to investigate the potential role of PRINS in diabetic nephropathy. The relative mRNA expression level of PRINS and SMAD family member 7 (Smad7) was examined in patients with diabetes, including patients without obvious complications (n=43), patients with diabetic nephropathy (n=33), diabetic retinopathy (n=37), diabetic cardiomyopathy (n=29), diabetic lung disease (n=38) and healthy controls (n=48). Correlation analysis between the expression level of PRINS and Smad7 was analyzed by Pearson's correlation analysis. In addition, overexpression of PRINS was confirmed in mouse podocyte cells and cell viability and Smad7 protein expression was detected by MTT assay and western blot analysis, respectively. The expression levels of PRINS and Smad7 were significantly increased in patients with diabetes compared with healthy controls. In addition, the expression levels of PRINS and Smad7 were significantly increased in patients with diabetic nephropathy compared with other diabetic complications. The expression level of PRINS in mouse podocyte cells was upregulated following treatment with high glucose. A significant positive correlation between the expression level of PRINS and Smad7 was observed in patients with diabetic nephropathy. However, there was no correlation was observed in other patient groups compared with healthy controls. Overexpression of PRINS decreased the viability of mouse podocyte cells and enhanced Smad7 protein expression. Taken together, these results suggest that PRINS may be involved in the development of nephropathy in patients with diabetes.
Collapse
Affiliation(s)
- Haiyan Jiao
- Department of Nephrology, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Daolin Xie
- Department of Ultrasound Diagnosis, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Yanhong Qiao
- Department of Nephrology, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| |
Collapse
|
50
|
May O, Merle NS, Grunenwald A, Gnemmi V, Leon J, Payet C, Robe-Rybkine T, Paule R, Delguste F, Satchell SC, Mathieson PW, Hazzan M, Boulanger E, Dimitrov JD, Fremeaux-Bacchi V, Frimat M, Roumenina LT. Heme Drives Susceptibility of Glomerular Endothelium to Complement Overactivation Due to Inefficient Upregulation of Heme Oxygenase-1. Front Immunol 2018; 9:3008. [PMID: 30619356 PMCID: PMC6306430 DOI: 10.3389/fimmu.2018.03008] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/05/2018] [Indexed: 11/27/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a severe disease characterized by microvascular endothelial cell (EC) lesions leading to thrombi formation, mechanical hemolysis and organ failure, predominantly renal. Complement system overactivation is a hallmark of aHUS. To investigate this selective susceptibility of the microvascular renal endothelium to complement attack and thrombotic microangiopathic lesions, we compared complement and cyto-protection markers on EC, from different vascular beds, in in vitro and in vivo models as well as in patients. No difference was observed for complement deposits or expression of complement and coagulation regulators between macrovascular and microvascular EC, either at resting state or after inflammatory challenge. After prolonged exposure to hemolysis-derived heme, higher C3 deposits were found on glomerular EC, in vitro and in vivo, compared with other EC in culture and in mice organs (liver, skin, brain, lungs and heart). This could be explained by a reduced complement regulation capacity due to weaker binding of Factor H and inefficient upregulation of thrombomodulin (TM). Microvascular EC also failed to upregulate the cytoprotective heme-degrading enzyme heme-oxygenase 1 (HO-1), normally induced by hemolysis products. Only HUVEC (Human Umbilical Vein EC) developed adaptation to heme, which was lost after inhibition of HO-1 activity. Interestingly, the expression of KLF2 and KLF4—known transcription factors of TM, also described as possible transcription modulators of HO-1- was weaker in micro than macrovascular EC under hemolytic conditions. Our results show that the microvascular EC, and especially glomerular EC, fail to adapt to the stress imposed by hemolysis and acquire a pro-coagulant and complement-activating phenotype. Together, these findings indicate that the vulnerability of glomerular EC to hemolysis is a key factor in aHUS, amplifying complement overactivation and thrombotic microangiopathic lesions.
Collapse
Affiliation(s)
- Olivia May
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,INSERM, UMR 995, Lille, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France
| | - Nicolas S Merle
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Grunenwald
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France.,University of Lille, INSERM, CHU Lille, Department of Pathology, UMR-S 1172 - Jean-Pierre Aubert Research Center, Lille, France
| | - Viviane Gnemmi
- University of Lille, INSERM, CHU Lille, Department of Pathology, UMR-S 1172 - Jean-Pierre Aubert Research Center, Lille, France
| | - Juliette Leon
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cloé Payet
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Tania Robe-Rybkine
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Romain Paule
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | - Marc Hazzan
- INSERM, UMR 995, Lille, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France
| | | | - Jordan D Dimitrov
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Veronique Fremeaux-Bacchi
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Marie Frimat
- INSERM, UMR 995, Lille, France.,University of Lille, CHU Lille, Nephrology Department, Lille, France
| | - Lubka T Roumenina
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|