1
|
Zhang S, Tang S, Liu Y, Xue B, Xie Q, Zhao L, Yuan H. Protein-bound uremic toxins as therapeutic targets for cardiovascular, kidney, and metabolic disorders. Front Endocrinol (Lausanne) 2025; 16:1500336. [PMID: 39931238 PMCID: PMC11808018 DOI: 10.3389/fendo.2025.1500336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/02/2025] [Indexed: 02/13/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome is a systemic clinical condition characterized by pathological and physiological interactions among metabolic abnormalities, chronic kidney disease, and cardiovascular diseases, leading to multi-organ dysfunction and a higher incidence of cardiovascular endpoints. Traditional approaches to managing CKM syndrome risk are inadequate in these patients, necessitating strategies targeting specific CKM syndrome risk factors. Increasing evidence suggests that addressing uremic toxins and/or pathways induced by uremic toxins may reduce CKM syndrome risk and treat the disease. This review explores the interactions among heart, kidney, and metabolic pathways in the context of uremic toxins and underscores the significant role of uremic toxins as potential therapeutic targets in the pathophysiology of these diseases. Strategies aimed at regulating these uremic toxins offer potential avenues for reversing and managing CKM syndrome, providing new insights for its clinical diagnosis and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huijuan Yuan
- Department of Endocrinology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Zhengzhou, China
| |
Collapse
|
2
|
Stanigut AM, Tuta L, Pana C, Alexandrescu L, Suceveanu A, Blebea NM, Vacaroiu IA. Autophagy and Mitophagy in Diabetic Kidney Disease-A Literature Review. Int J Mol Sci 2025; 26:806. [PMID: 39859520 PMCID: PMC11766107 DOI: 10.3390/ijms26020806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Autophagy and mitophagy are critical cellular processes that maintain homeostasis by removing damaged organelles and promoting cellular survival under stress conditions. In the context of diabetic kidney disease, these mechanisms play essential roles in mitigating cellular damage. This review provides an in-depth analysis of the recent literature on the relationship between autophagy, mitophagy, and diabetic kidney disease, highlighting the current state of knowledge, existing research gaps, and potential areas for future investigations. Diabetic nephropathy (DN) is traditionally defined as a specific form of kidney disease caused by long-standing diabetes, characterized by the classic histological lesions in the kidney, including mesangial expansion, glomerular basement membrane thickening, nodular glomerulosclerosis (Kimmelstiel-Wilson nodules), and podocyte injury. Clinical markers for DN are albuminuria and the gradual decline in glomerular filtration rate (GFR). Diabetic kidney disease (DKD) is a broader and more inclusive term, for all forms of chronic kidney disease (CKD) in individuals with diabetes, regardless of the underlying pathology. This includes patients who may have diabetes-associated kidney damage without the typical histological findings of diabetic nephropathy. It also accounts for patients with other coexisting kidney diseases (e.g., hypertensive nephrosclerosis, ischemic nephropathy, tubulointerstitial nephropathies), even in the absence of albuminuria, such as a reduction in GFR.
Collapse
Affiliation(s)
- Alina Mihaela Stanigut
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Nephrology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Liliana Tuta
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Nephrology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Camelia Pana
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Nephrology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Luana Alexandrescu
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Gastroenterology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Adrian Suceveanu
- Clinical Medical Disciplines Department, Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.M.S.); (L.T.); (L.A.); (A.S.)
- Gastroenterology Department, County Emergency Clinical Hospital of Constanta, 145 Tomis Street, 900591 Constanta, Romania
| | - Nicoleta-Mirela Blebea
- Department of Pharmacotherapy, Faculty of Pharmacy, Ovidius University of Constanta, Aleea Universitatii Nr. 1, 900470 Constanta, Romania
| | - Ileana Adela Vacaroiu
- Department of Nephrology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Nephrology, Sf. Ioan Clinical Emergency Hospital, 042122 Bucharest, Romania
| |
Collapse
|
3
|
Liu WC, Wu MY, Lim PS. Alterations in Autophagic Function and Endoplasmic Reticulum Stress Markers in the Peripheral Blood Mononuclear Cells of Patients on Hemodialysis. Int J Mol Sci 2025; 26:447. [PMID: 39859163 PMCID: PMC11764487 DOI: 10.3390/ijms26020447] [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: 12/07/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Oxidative stress, endoplasmic reticulum (ER) stress, and alterations in autophagy activity have been described as prominent factors mediating many pathological processes in chronic kidney disease (CKD). The accumulation of misfolded proteins in the ER may stimulate the unfolded protein response (UPR). The interplay between autophagy and UPR in hemodialysis (HD) patients remains unclear. The aim of the present study was to explore the associations between serum oxidative stress markers, autophagy activity, and ER stress markers in the peripheral blood mononuclear cells (PBMCs) of patients on HD. Autophagy and ER stress-related protein expression levels in PBMCs were measured using western blotting. The redox state of human serum albumin was measured via high-performance liquid chromatography. Levels of the microtubule associated protein light chain 3 (LC3)-II, BECLIN1, and p62/SQSTM1 proteins were significantly increased in PBMCs of HD patients compared to healthy subjects. The PBMCs in HD patients also displayed augmented glucose-regulated protein 78 kDa (GRP78), phosphorylated eukaryotic translation initiation factor 2, subunit 1 alpha (p-eIF2α), and activating transcription factor 6 (ATF6) levels (p < 0.05). Additionally, nuclear factor erythroid 2 (NF-E2)-related factor 2 (NRF2) levels were elevated in the PBMCs of HD patients, compared to those of healthy subjects. Correlation analysis showed that the redox status of albumin was significantly correlated with the p62 protein level in PBMCs. Compared to healthy controls, we found elevated autophagosome formation in HD patients. Increased expression of ER stress markers was also observed in HD patients. Furthermore, increased p62 expression was positively correlated with the protein expression of NRF2, as well as a reduced form of serum albumin (human mercaptalbumin; HMA), in HD patients.
Collapse
Affiliation(s)
- Wen-Chih Liu
- Section of Nephrology, Department of Medicine, Antai Medical Care Corporation Antai Tian-Sheng Memorial Hospital, Pingtung 928, Taiwan;
- Department of Nursing, Meiho University, Pingtung 912, Taiwan
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan
| | - Ming-Yin Wu
- Division of Renal Medicine, Tungs’ Taichung MetroHarbor Hospital, Taichung 433, Taiwan;
| | - Paik Seong Lim
- Division of Renal Medicine, Tungs’ Taichung MetroHarbor Hospital, Taichung 433, Taiwan;
- Institute of Biomedical Science, College of Life Science, National Chung Hsing University, Taiwan Hospital, Taichung 402, Taiwan
| |
Collapse
|
4
|
Hebah HA, Kamel HM, Bastawy IM, Ahmed FA. Association between serum beclin 1 level and cardiac valvular calcification in hemodialysis patients. Curr Probl Cardiol 2024; 49:102519. [PMID: 38492612 DOI: 10.1016/j.cpcardiol.2024.102519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Cardiovascular calcification is a pervasive issue throughout chronic kidney disease (CKD) progression. Autophagy, a fundamental cellular process, exerts significant influence on various cardiac pathologies, including arrhythmias, atherosclerosis, heart failure, and notably, valvular, and vascular calcifications. Beclin-1, a crucial eukaryotic protein, plays a major regulatory role in autophagy as part of the phosphatidylinositol-3-kinase (PI3K) complex. Recent evidence suggests a protective role for Beclin-1-mediated autophagy in CKD vascular calcification, raising its potential as a novel therapeutic target in this context. WE AIMED TO Investigate the association between serum Beclin 1 levels and the presence of cardiovascular valvular calcification in hemodialysis patients. RESULTS This study evaluated a cohort of 102 hemodialysis patients, evenly divided into two groups based on echocardiographic findings. All participants underwent serum Beclin 1 measurement and transthoracic echocardiography. Patients with acute kidney injury, active malignancy, or diabetes were excluded. Our study revealed significant differences between the two groups in terms of: Serum Beclin 1 levels, all parameters of lipid profile, prevalence of ischemic heart disease, serum albumin levels and Total calcium. Echocardiography in Group 1 showed that most cases (60.78%) exhibited mild aortic valve calcification. Additionally, significant relationships were observed between Beclin 1 and: ischemic heart disease (p=0.011) Aortic valve calcification on echocardiography (p < 0.001) Interestingly, lower Beclin 1 levels were associated with more severe valve calcification. A Beclin 1 cutoff value of ≤ 35.5 ng/ml demonstrated the highest sensitivity (98%) and specificity (92%). CONCLUSION Our findings suggest that the serum Beclin 1 level could be incorporated into a predictive model for cardiac valvular calcification in hemodialysis patients.
Collapse
Affiliation(s)
- Hayam Ahmed Hebah
- Ain Shams University, Faculty of Medicine, Internal Medicine, And Nephrology Department. Cairo, Egypt
| | - Hadeer Moamen Kamel
- Ain Shams University, Faculty of Medicine, Internal Medicine, And Nephrology Department. Cairo, Egypt
| | | | - Fatma Abdelrahman Ahmed
- Ain Shams University, Faculty of Medicine, Internal Medicine, And Nephrology Department. Cairo, Egypt.
| |
Collapse
|
5
|
Granata S, Bruschi M, Verlato A, Pontrelli P, Gesualdo L, Stallone G, Zaza G. Autophagy Activation in Peripheral Blood Mononuclear Cells of Peritoneal Dialysis Patients. Kidney Int Rep 2023; 8:1852-1863. [PMID: 37705917 PMCID: PMC10496084 DOI: 10.1016/j.ekir.2023.06.017] [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: 03/22/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 09/15/2023] Open
Abstract
Introduction The complete systemic deregulated biological network in patients on peritoneal dialysis (PD) is still only partially defined. High-throughput/omics techniques may offer the possibility to analyze the main biological fingerprints associated with this clinical condition. Methods We applied an innovative bioinformatic analysis of gene expression microarray data (mainly based on support vector machine (SVM) learning) to compare the transcriptomic profile of peripheral blood mononuclear cells (PBMCs) of healthy subjects (HS), chronic kidney disease (CKD) patients, and patients on PD divided into a microarray group (5 HS, 9 CKD, and 10 PD) and a validation group (10 HS, 15 CKD, and 15 PD). Classical well-standardized biomolecular approaches (western blotting and flow cytometry) were used to validate the transcriptomic results. Results Bioinformatics revealed a distinctive PBMC transcriptomic profiling for PD versus CKD and HS (n = 419 genes). Transcripts encoding for key elements of the autophagic pathway were significantly upregulated in PD, and the autophagy related 5 (ATG5) reached the top level of discrimination [-Log10 P-value = 11.3, variable importance in projection (VIP) score = 4.8, SVM rank:1]. Protein levels of ATG5 and microtubule associated protein 1 light chain 3 beta (LC3B), an important constituent of the autophagosome, validated microarray results. In addition, the incubation of PBMCs of HS with serum of patients on PD upregulated both proteins. Autophagy in PBMCs from patients on PD was attenuated by N-acetyl-cysteine or Resatorvid treatment. Conclusions Our data demonstrated, for the first time, that the autophagy pathway is activated in immune-cells of patients on PD, and this may represent a novel therapeutic target.
Collapse
Affiliation(s)
- Simona Granata
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
- Department of Medicine, Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gianluigi Zaza
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
6
|
Ranjbaran M, Kadkhodaee M, Adelipour M, Hafazeh L, Lorian K, Seifi B. A comparison between centrally and systemically administered erythropoietin on kidney protection in a model of fixed-volume hemorrhagic shock in male rats. Mol Biol Rep 2023; 50:4781-4789. [PMID: 37024748 DOI: 10.1007/s11033-023-08412-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND In this study, a comparison between centrally and systemically administered erythropoietin (EPO) was performed on nephroprotection during hemorrhagic shock (HS) in male rats. METHODS Male rats were allocated into four experimental groups. (1) Sham; a guide cannula was inserted into the left lateral ventricle and other cannulas were placed into the left femoral artery and vein. (2) HS; stereotaxic surgery was done to insert a cannula in the left lateral ventricle and after a 7-day recovery; hemorrhagic shock and resuscitation were performed. (3) EPO-systemic; the procedure was the same as the HS group except that animals received 300 IU/kg erythropoietin into the femoral vein immediately before resuscitation. (4) EPO-central; animals was treated with erythropoietin (2 IU/rat) into the left lateral ventricle before resuscitation. Arterial oxygen saturation (SaO2) was measured during experiments. Urine and renal tissue samples were stored for ex-vivo indices assessments. RESULTS Erythropoietin (systemically/centrally administered) significantly improved SaO2, renal functional and oxidative stress parameters and decreased renal inflammatory (TNF-α and IL-6) mRNA expression compared to the HS group. EPO-treated groups showed a decrease in active form of caspase-3 protein level and an increase in autophagy activity in comparison with the HS group. CONCLUSION Considering the fact that the effective dose of systemic EPO (300 IU/kg) was roughly 50 times higher than that of central administration (2 IU/rat), centrally administered EPO was accompanied by more advantageous consequences than systemic way. EPO is likely to act as a neuro-modulator or neuro-mediator in the central protection of organs including the kidneys.
Collapse
Affiliation(s)
- Mina Ranjbaran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehri Kadkhodaee
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Adelipour
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Hafazeh
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keivan Lorian
- Research and clinical center for infertility, Yazd Rreproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Behjat Seifi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Priego AR, Parra EG, Mas S, Morgado-Pascual JL, Ruiz-Ortega M, Rayego-Mateos S. Bisphenol A Modulates Autophagy and Exacerbates Chronic Kidney Damage in Mice. Int J Mol Sci 2021; 22:7189. [PMID: 34281243 PMCID: PMC8268806 DOI: 10.3390/ijms22137189] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bisphenol A (BPA) is a ubiquitous environmental toxin that accumulates in chronic kidney disease (CKD). Our aim was to explore the effect of chronic exposition of BPA in healthy and injured kidney investigating potential mechanisms involved. METHODS In C57Bl/6 mice, administration of BPA (120 mg/kg/day, i.p for 5 days/week) was done for 2 and 5 weeks. To study BPA effect on CKD, a model of subtotal nephrectomy (SNX) combined with BPA administration for 5 weeks was employed. In vitro studies were done in human proximal tubular epithelial cells (HK-2 line). RESULTS Chronic BPA administration to healthy mice induces inflammatory infiltration in the kidney, tubular injury and renal fibrosis (assessed by increased collagen deposition). Moreover, in SNX mice BPA exposure exacerbates renal lesions, including overexpression of the tubular damage biomarker Hepatitis A virus cellular receptor 1 (Havcr-1/KIM-1). BPA upregulated several proinflammatory genes and increased the antioxidant response [Nuclear factor erythroid 2-related factor 2 (Nrf2), Heme Oxygenase-1 (Ho-1) and NAD(P)H dehydrogenase quinone 1 (Nqo-1)] both in healthy and SNX mice. The autophagy process was modulated by BPA, through elevated autophagy-related gene 5 (Atg5), autophagy-related gene 7 (Atg7), Microtubule-associated proteins 1A/1B light chain 3B (Map1lc3b/Lc3b) and Beclin-1 gene levels and blockaded the autophagosome maturation and flux (p62 levels). This autophagy deregulation was confirmed in vitro. CONCLUSIONS BPA deregulates autophagy flux and redox protective mechanisms, suggesting a potential mechanism of BPA deleterious effects in the kidney.
Collapse
Affiliation(s)
- Alberto Ruiz Priego
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-UAM/IRSIN, 28040 Madrid, Spain; (A.R.P.); (S.M.)
| | - Emilio González Parra
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-UAM/IRSIN, 28040 Madrid, Spain; (A.R.P.); (S.M.)
| | - Sebastián Mas
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-UAM/IRSIN, 28040 Madrid, Spain; (A.R.P.); (S.M.)
| | - José Luis Morgado-Pascual
- Cellular Biology, Physiology and Immunology Department, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain;
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid Faculty of Medicine, 28040 Madrid, Spain;
| | - Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid Faculty of Medicine, 28040 Madrid, Spain;
| |
Collapse
|
8
|
Guan X, Shen J, Xu Y, Feng X, Zhou R. Heme oxygenase-1 enhances autophagy by modulating the AMPK/mTORC1 signaling pathway as a renoprotective mechanism to mitigate lead-induced nephrotoxicity. Am J Transl Res 2020; 12:4807-4818. [PMID: 32913552 PMCID: PMC7476133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
Lead (Pb), a highly poisonous heavy metal and an important occupational hazard, is currently a widespread environmental pollutant. The kidney is especially susceptible to the toxic effects of Pb because of its major role in Pb excretion. Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that can mitigate cellular injury. However, its role in Pb-elicited nephrotoxicity remains uncertain. This study was designed to examine the role of HO-1 in lead acetate (PbAc)-induced renal tubular cell injury in vitro. PbAc injury was found to suppress HO-1 expression and impair cell viability, with concomitant depletion of the autophagy proteins LC3-II and Beclin 1. Overexpression of HO-1 dramatically restored autophagy and protected cells against PbAc-induced apoptosis. In addition, pretreatment with 3-methyladenine, an inhibitor of autophagy, aggravated apoptosis and abolished renoprotection by HO-1, suggesting that the anti-apoptotic effect of HO-1 in Pb-induced nephrotoxicity is dependent on enhanced autophagy. Furthermore, HO-1 overexpression abrogated the inhibitory effect of PbAc on the adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTORC1) signaling pathway. Pretreatment with an AMPK agonist, 5-aminoimidazole-4-carboxamide-1-β-D ribofuranoside, markedly enhanced autophagic activity and diminished apoptosis. Conversely, inhibition of AMPK phosphorylation abolished the pro-autophagic and anti-apoptotic effects of HO-1 in PbAc-injured cells. Our findings suggest that HO-1 alleviates Pb-induced nephrotoxicity via enhanced autophagy, which involves activation of the AMPK/mTORC1 signaling pathway.
Collapse
Affiliation(s)
- Xuejing Guan
- Department of Nephrology, Yangpu Hospital, Tongji University School of Medicine Shanghai, China
| | - Jie Shen
- Department of Nephrology, Yangpu Hospital, Tongji University School of Medicine Shanghai, China
| | - Yahong Xu
- Department of Nephrology, Yangpu Hospital, Tongji University School of Medicine Shanghai, China
| | - Xuefang Feng
- Department of Nephrology, Yangpu Hospital, Tongji University School of Medicine Shanghai, China
| | - Rong Zhou
- Department of Nephrology, Yangpu Hospital, Tongji University School of Medicine Shanghai, China
| |
Collapse
|
9
|
Abstract
Cardiovascular disease and infections are major causes for the high incidence of morbidity and mortality of patients with chronic kidney disease. Both complications are directly or indirectly associated with disturbed functions or altered apoptotic rates of polymorphonuclear leukocytes, monocytes, lymphocytes, and dendritic cells. Normal responses of immune cells can be reduced, leading to infectious diseases or pre-activated/primed, giving rise to inflammation and subsequently to cardiovascular disease. This review summarizes the impact of kidney dysfunction on the immune system. Renal failure results in disturbed renal metabolic activities with reduced renin, erythropoietin, and vitamin D production, which adversely affects the immune system. Decreased kidney function also leads to reduced glomerular filtration and the retention of uremic toxins. A large number of uremic toxins with detrimental effects on immune cells have been identified. Besides small water-soluble and protein-bound compounds originating from the intestinal microbiome, several molecules in the middle molecular range, e.g., immunoglobulin light chains, retinol-binding protein, the neuropeptides Met-enkephalin and neuropeptide Y, endothelin-1, and the adipokines leptin and resistin, adversely affect immune cells. Posttranslational modifications such as carbamoylation, advanced glycation products, and oxidative modifications contribute to uremic toxicity. Furthermore, high-density lipoprotein from uremic patients has an altered protein profile and thereby loses its anti-inflammatory properties.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
10
|
De Marañon AM, Iannantuoni F, Abad-Jiménez Z, Canet F, Díaz-Pozo P, López-Domènech S, Jover A, Morillas C, Mariño G, Apostolova N, Rocha M, Victor VM. Relationship between PMN-endothelium interactions, ROS production and Beclin-1 in type 2 diabetes. Redox Biol 2020; 34:101563. [PMID: 32416353 PMCID: PMC7226867 DOI: 10.1016/j.redox.2020.101563] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes is closely related to oxidative stress and cardiovascular diseases. In this study, we hypothesized that polymorphonuclear leukocytes (PMN)-endothelium interactions and autophagy are associated. We evaluated PMN-endothelial interactions, ROS production and autophagy parameters in 47 type 2 diabetic patients and 57 control subjects. PMNs from type 2 diabetic patients exhibited slower rolling velocity (p < 0.001), higher rolling flux (p < 0.001) and adhesion (p < 0.001) in parallel to higher levels of total (p < 0.05) and mitochondrial ROS (p < 0.05). When the protein expression of autophagy markers was analysed, an increase of Beclin-1 (p < 0.05), LC3I (p < 0.05), LC3II (p < 0.01) and LC3II/LC3I ratio (p < 0.05) was observed. Several correlations between ROS and leukocyte-endothelium parameters were found. Interestingly, in control subjects, an increase of Beclin-1 levels was accompanied by a decrease in the number of rolling (r = 0.561) and adhering PMNs (r = 0.560) and a rise in the velocity of the rolling PMNs (r = 0.593). In contrast, in the type 2 diabetic population, a rise in Beclin-1 levels was related to an increase in the number of rolling (r = 0.437), and adhering PMNs (r = 0.467). These results support the hypothesis that PMN-endothelium interactions, ROS levels and formation of autophagosomes, especially Beclin-1 levels, are enhanced in type 2 diabetes.
Collapse
Affiliation(s)
- Aranzazu M De Marañon
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Francesca Iannantuoni
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Zaida Abad-Jiménez
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Francisco Canet
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Pedro Díaz-Pozo
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Sandra López-Domènech
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Ana Jover
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Carlos Morillas
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Guillermo Mariño
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011, Oviedo, Spain
| | - Nadezda Apostolova
- CIBERehd - Department of Pharmacology, University of Valencia, Valencia, Spain
| | - Milagros Rocha
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; CIBERehd - Department of Pharmacology, University of Valencia, Valencia, Spain.
| | - Victor M Victor
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; CIBERehd - Department of Pharmacology, University of Valencia, Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain.
| |
Collapse
|
11
|
Wang YJ, Chen YY, Hsiao CM, Pan MH, Wang BJ, Chen YC, Ho CT, Huang KC, Chen RJ. Induction of Autophagy by Pterostilbene Contributes to the Prevention of Renal Fibrosis via Attenuating NLRP3 Inflammasome Activation and Epithelial-Mesenchymal Transition. Front Cell Dev Biol 2020; 8:436. [PMID: 32582712 PMCID: PMC7283393 DOI: 10.3389/fcell.2020.00436] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/08/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) is recognized as a global public health problem. NLRP3 inflammasome activation has been characterized to mediate diverse aspect mechanisms of CKD through regulation of proinflammatory cytokines, tubulointerstitial injury, glomerular diseases, renal inflammation, and fibrosis pathways. Autophagy is a characterized negative regulation mechanism in the regulation of the NLRP3 inflammasome, which is now recognized as the key regulator in the pathogenesis of inflammation and fibrosis in CKD. Thus, autophagy is undoubtedly an attractive target for developing new renal protective treatments of kidney disease via its potential effects in regulation of inflammasome. However, there is no clinical useful agent targeting the autophagy pathway for patients with renal diseases. Pterostilbene (PT, trans-3,5-dimethoxy-4-hydroxystilbene) is a natural analog of resveratrol that has various health benefits including autophagy inducing effects. Accordingly, we aim to investigate underlying mechanisms of preventive and therapeutic effects of PT by reducing NLRP3 inflammasome activation and fibrosis through autophagy-inducing effects. The renal protective effects of PT were evaluated by potassium oxonate (PO)-induced hyperuricemia and high adenine diet-induced CKD models. The autophagy induction mechanisms and anti-fibrosis effects of PT by down-regulation of NLRP3 inflammasome are investigated by using immortalized rat kidney proximal tubular epithelial NRK-52E cells. To determine the role of autophagy induction in the alleviating of NLRP3 inflammasome activation and epithelial-mesenchymal transition (EMT), NRK-52E with Atg5 knockdown [NRK-Atg5-(2)] cells were applied in the study. The results indicated that PT significantly reduces serum uric acid levels, liver xanthine oxidase activity, collagen accumulation, macrophage recruitment, and renal fibrosis in CKD models. At the molecular levels, pretreatment with PT downregulating TGF-β-triggered NLRP3 inflammasome activation, and subsequent EMT in NRK-52E cells. After blockage of autophagy by treatment of Atg5 shRNA, PT loss of its ability to prevent NLRP3 inflammasome activation and EMT. Taken together, we suggested the renal protective effects of PT in urate nephropathy and proved that PT induces autophagy leading to restraining TGF-β-mediated NLRP3 inflammasome activation and EMT. This study is also the first one to provide a clinical potential application of PT for a better management of CKD through its autophagy inducing effects.
Collapse
Affiliation(s)
- Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Mao Hsiao
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Bour-Jr Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.,Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Yu-Chi Chen
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, United States
| | - Kuo-Ching Huang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Division of Nephrology, Department of Internal Medicine, Chi Mei Hospital, Tainan, Taiwan
| | - Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
12
|
Wang CY, Lin TA, Ho MY, Yeh JK, Tsai ML, Hung KC, Hsieh IC, Wen MS. Regulation of autophagy in leukocytes through RNA N 6-adenosine methylation in chronic kidney disease patients. Biochem Biophys Res Commun 2020; 527:953-959. [PMID: 32439179 DOI: 10.1016/j.bbrc.2020.04.138] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022]
Abstract
Patients with chronic kidney diseases have multiple cellular dysfunctions leading to increased atherosclerosis, impaired immunity, and disturbed metabolism. However, it is unclear what is the fundamental signaling served as a marker or as a mediator for the dysregulated function in their leukocytes or tissues. Here we hypothesized that the N6-Methyladenosine (m6A) modification of the RNA in the leukocytes is responsible for the cellular dysfunction in chronic kidney diseases. Patients with chronic kidney diseases had significantly less m6A abundances in leukocytes and elevated RNA demethylase FTO proteins. The uremic toxin, indoxyl sulfate, activated the autophagy flux through modulation of FTO and m6A modifications in RNA. Notably, knockdown of FTO or inhibit the m6A by 3-deazaadenosine blocks the effects of indoxyl sulfate on autophagy activation in cells. These findings provide new insights into the mechanisms underlying chronic kidney disease-associated cellular dysfunction. Targeting RNA m6A modification may be a novel strategy for the treatment of chronic kidney diseases and autophagy.
Collapse
Affiliation(s)
- Chao-Yung Wang
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan; Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, 350, Taiwan.
| | - Tien-An Lin
- Department of General Surgery, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan
| | - Ming-Yun Ho
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan
| | - Jih-Kai Yeh
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan
| | - Ming-Lung Tsai
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan
| | - Kuo-Chun Hung
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan
| | - I-Chang Hsieh
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan
| | - Ming-Shien Wen
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taiwan
| |
Collapse
|
13
|
Rodrigues SD, Santos SS, Meireles T, Romero N, Glorieux G, Pecoits-Filho R, Zhang DD, Nakao LS. Uremic toxins promote accumulation of oxidized protein and increased sensitivity to hydrogen peroxide in endothelial cells by impairing the autophagic flux. Biochem Biophys Res Commun 2019; 523:123-129. [PMID: 31837804 DOI: 10.1016/j.bbrc.2019.12.022] [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: 11/09/2019] [Accepted: 12/05/2019] [Indexed: 01/12/2023]
Abstract
Chronic kidney disease (CKD) is associated with high mortality rates, mainly due to cardiovascular diseases (CVD). Uremia has been considered a relevant risk factor for CVD in CKD patients, since uremic toxins (UTs) promote systemic and vascular inflammation, oxidative stress and senescence. Here, we demonstrate that uremic toxins indoxyl sulfate (IxS), p-cresyl sulfate (pCS) and indole acetic acid (IAA) are incorporated by human endothelial cells and inhibit the autophagic flux, demonstrated by cellular p62 accumulation. Moreover, isolated and mixed UTs impair the lysosomal stage of autophagy, as determined by cell imaging of the mRFP-GFP-LC3 protein. Endothelial cells exposed to UTs display accumulation of carbonylated proteins and increased sensitivity to hydrogen peroxide. Rapamycin, an autophagy activator which induces both autophagosome formation and clearance, prevented these effects. Collectively, our findings demonstrate that accumulation of oxidized proteins and enhanced cell sensitivity to hydrogen peroxide are consequences of impaired autophagic flux. These data provide evidence that UTs-induced impaired autophagy may be a novel contributor to endothelial dysfunction.
Collapse
Affiliation(s)
- Silvia D Rodrigues
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil
| | - Sabrina S Santos
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil
| | - Tassiana Meireles
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil
| | - Natalia Romero
- Cell Analysis Division, Agilent Technologies, 02421, Lexington, MA, USA
| | - Griet Glorieux
- Department of Nephrology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Roberto Pecoits-Filho
- Center for Health and Biological Sciences, Pontific Catholic University of Paraná, 80215-901, Curitiba, Brazil
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, University of Arizona, 85721, Tucson, AZ, USA
| | - Lia S Nakao
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil.
| |
Collapse
|
14
|
Autophagy in Chronic Kidney Diseases. Cells 2019; 8:cells8010061. [PMID: 30654583 PMCID: PMC6357204 DOI: 10.3390/cells8010061] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a cellular recycling process involving self-degradation and reconstruction of damaged organelles and proteins. Current evidence suggests that autophagy is critical in kidney physiology and homeostasis. In clinical studies, autophagy activations and inhibitions are linked to acute kidney injuries, chronic kidney diseases, diabetic nephropathies, and polycystic kidney diseases. Oxidative stress, inflammation, and mitochondrial dysfunction, which are implicated as important mechanisms underlying many kidney diseases, modulate the autophagy activation and inhibition and lead to cellular recycling dysfunction. Abnormal autophagy function can induce loss of podocytes, damage proximal tubular cells, and glomerulosclerosis. After acute kidney injuries, activated autophagy protects tubular cells from apoptosis and enhances cellular regeneration. Patients with chronic kidney diseases have impaired autophagy that cannot be reversed by hemodialysis. Multiple nephrotoxic medications also alter the autophagy signaling, by which the mechanistic insights of the drugs are revealed, thus providing the unique opportunity to manage the nephrotoxicity of these drugs. In this review, we summarize the current concepts of autophagy and its molecular aspects in different kidney cells pathophysiology. We also discuss the current evidence of autophagy in acute kidney injury, chronic kidney disease, toxic effects of drugs, and aging kidneys. In addition, we examine therapeutic possibilities targeting the autophagy system in kidney diseases.
Collapse
|
15
|
Guo H, Wang B, Li H, Ling L, Niu J, Gu Y. Glucagon-like peptide-1 analog prevents obesity-related glomerulopathy by inhibiting excessive autophagy in podocytes. Am J Physiol Renal Physiol 2017; 314:F181-F189. [PMID: 29070572 DOI: 10.1152/ajprenal.00302.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To investigate the role of glucagon-like peptide-1 analog (GLP-1) in high-fat diet-induced obesity-related glomerulopathy (ORG). Male C57BL/6 mice fed a high-fat diet for 12 wk were treated with GLP-1 (200 μg/kg) or 0.9% saline for 4 wk. Fasting blood glucose and insulin and the expression of podocin, nephrin, phosphoinositide 3-kinase (PI3K), glucose transporter type (Glut4), and microtubule-associated protein 1A/1B-light chain 3 (LC3) were assayed. Glomerular morphology and podocyte foot structure were evaluated by periodic acid-Schiff staining and electron microscopy. Podocytes were treated with 150 nM GLP-1 and incubated with 400 μM palmitic acid (PA) for 12 h. The effect on autophagy was assessed by podocyte-specific Glut4 siRNA. Insulin resistance and autophagy were assayed by immunofluorescence and Western blotting. The high-fat diet resulted in weight gain, ectopic glomerular lipid accumulation, increased insulin resistance, and fusion of podophyte foot processes. The decreased translocation of Glut4 to the plasma membrane and excess autophagy seen in mice fed a high-fat diet and in PA-treated cultured podocytes were attenuated by GLP-1. Podocyte-specific Glut4 siRNA promoted autophagy, and rapamycin-enhanced autophagy worsened the podocyte injury caused by PA. Excess autophagy in podocytes was induced by inhibition of Glut4 translocation to the plasma membrane and was involved in the pathology of ORG. GLP-1 restored insulin sensitivity and ameliorated renal injury by decreasing the level of autophagy.
Collapse
Affiliation(s)
- Honglei Guo
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China.,Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu Province, China
| | - Bin Wang
- Department of Nephrology, Zhongda Hospital Southeast University, Nanjing, Jiangsu Province, China
| | - Hongmei Li
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Lilu Ling
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Jianying Niu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Yong Gu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China.,Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University , Shanghai , China
| |
Collapse
|
16
|
Kooman JP, Dekker MJ, Usvyat LA, Kotanko P, van der Sande FM, Schalkwijk CG, Shiels PG, Stenvinkel P. Inflammation and premature aging in advanced chronic kidney disease. Am J Physiol Renal Physiol 2017; 313:F938-F950. [PMID: 28701312 DOI: 10.1152/ajprenal.00256.2017] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/29/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022] Open
Abstract
Systemic inflammation in end-stage renal disease is an established risk factor for mortality and a catalyst for other complications, which are related to a premature aging phenotype, including muscle wasting, vascular calcification, and other forms of premature vascular disease, depression, osteoporosis, and frailty. Uremic inflammation is also mechanistically related to mechanisms involved in the aging process, such as telomere shortening, mitochondrial dysfunction, and altered nutrient sensing, which can have a direct effect on cellular and tissue function. In addition to uremia-specific causes, such as abnormalities in the phosphate-Klotho axis, there are remarkable similarities between the pathophysiology of uremic inflammation and so-called "inflammaging" in the general population. Potentially relevant, but still somewhat unexplored in this respect, are abnormal or misplaced protein structures, as well as abnormalities in tissue homeostasis, which evoke danger signals through damage-associated molecular patterns, as well as the senescence-associated secretory phenotype. Systemic inflammation, in combination with the loss of kidney function, can impair the resilience of the body to external and internal stressors by reduced functional and structural tissue reserves, and by impairing normal organ crosstalk, thus providing an explanation for the greatly increased risk of homeostatic breakdown in this population. In this review, the relationship between uremic inflammation and a premature aging phenotype, as well as potential causes and consequences, are discussed.
Collapse
Affiliation(s)
- Jeroen P Kooman
- Maastricht University Medical Center, Maastricht, Netherlands;
| | | | - Len A Usvyat
- Fresenius Medical Care North America, Waltham, Massachusetts
| | - Peter Kotanko
- Renal Research Institute, New York, New York.,Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Paul G Shiels
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; and
| | - Peter Stenvinkel
- Divsion of Renal Medicine, Department of Clinical Science Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Abstract
Many common renal insults such as ischemia and toxic injury primarily target the tubular epithelial cells, especially the highly metabolically active proximal tubular segment. Tubular epithelial cells are particularly dependent on autophagy to maintain homeostasis and respond to stressors. The pattern of autophagy in the kidney has a unique spatial and chronologic signature. Recent evidence has shown that there is complex cross-talk between autophagy and various cell death pathways. This review specifically discusses the interplay between autophagy and cell death in the renal tubular epithelia. It is imperative to review this topic because recent discoveries have improved our mechanistic understanding of the autophagic process and have highlighted its broad clinical applications, making autophagy a major target for drug development.
Collapse
Affiliation(s)
- Andrea Havasi
- Department of Nephrology, Boston University Medical Center, Boston, MA.
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia and Charlie Norwood VA Medical Center, Augusta, GA
| |
Collapse
|
18
|
Kooman JP, Shiels PG, Stenvinkel P. Premature aging in chronic kidney disease and chronic obstructive pulmonary disease: similarities and differences. Curr Opin Clin Nutr Metab Care 2015; 18:528-34. [PMID: 26372510 DOI: 10.1097/mco.0000000000000218] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW There is increasing clinical and pathophysiological evidence that a premature aging process is involved in the pathogenesis of systemic complications of many chronic organ diseases, which result in analogous phenotypes, including premature vascular aging, osteoporosis and muscle wasting. Novel developments from research into the aging process will, therefore, have relevance for understanding complications of organ diseases, such as chronic kidney disease and chronic obstructive pulmonary disease. The aim of the present article is to combine recent literature on aging mechanisms with evidence on the pathogenesis of systemic complications of these two chronic debilitating disorders. RECENT FINDINGS Recently, nine hallmarks of aging have been identified. In this review, we argue that all of these hallmarks are relevant for the pathogenesis of premature aging processes in chronic obstructive pulmonary disease and chronic kidney disease. Additionally, organ-specific alterations in proaging mechanisms, which reveal differences in phenotype against a generic background of premature aging, will be addressed. However, within patient populations who share a common diagnosis, clusters of patients with different phenotypes may be identified, which may show overlap with patients with other chronic diseases. SUMMARY An increased understanding of the premature aging process as well as its systemic consequences may pave the way for 'precision' intervention as well as shared treatment opportunities between chronic debilitating diseases of various causes.
Collapse
Affiliation(s)
- Jeroen P Kooman
- aDivision of Nephrology, Department of Internal Medicine, University Hospital Maastricht, Maastricht, the Netherlands bInstitute of Cancer Sciences, Wolfson Wohl Translational Research Centre, University of Glasgow, Glasgow, UK cDivision of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Stockholm, Sweden
| | | | | |
Collapse
|
19
|
White WE, Yaqoob MM, Harwood SM. Aging and uremia: Is there cellular and molecular crossover? World J Nephrol 2015; 4:19-30. [PMID: 25664244 PMCID: PMC4317625 DOI: 10.5527/wjn.v4.i1.19] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/28/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Many observers have noted that the morphological changes that occur in chronic kidney disease (CKD) patients resemble those seen in the geriatric population, with strikingly similar morbidity and mortality profiles and rates of frailty in the two groups, and shared characteristics at a pathophysiological level especially in respect to the changes seen in their vascular and immune systems. However, whilst much has been documented about the shared physical characteristics of aging and uremia, the molecular and cellular similarities between the two have received less attention. In order to bridge this perceived gap we have reviewed published research concerning the common molecular processes seen in aging subjects and CKD patients, with specific attention to altered proteostasis, mitochondrial dysfunction, post-translational protein modification, and senescence and telomere attrition. We have also sought to illustrate how the cell death and survival pathways apoptosis, necroptosis and autophagy are closely interrelated, and how an understanding of these overlapping pathways is helpful in order to appreciate the shared molecular basis behind the pathophysiology of aging and uremia. This analysis revealed many common molecular characteristics and showed similar patterns of cellular dysfunction. We conclude that the accelerated aging seen in patients with CKD is underpinned at the molecular level, and that a greater understanding of these molecular processes might eventually lead to new much needed therapeutic strategies of benefit to patients with renal disease.
Collapse
|