1
|
Asadi R, Shadpour P, Nakhaei A. Non-dialyzable uremic toxins and renal tubular cell damage in CKD patients: a systems biology approach. Eur J Med Res 2024; 29:412. [PMID: 39123228 PMCID: PMC11311939 DOI: 10.1186/s40001-024-01951-z] [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: 08/02/2023] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Chronic kidney disease presents global health challenges, with hemodialysis as a common treatment. However, non-dialyzable uremic toxins demand further investigation for new therapeutic approaches. Renal tubular cells require scrutiny due to their vulnerability to uremic toxins. METHODS In this study, a systems biology approach utilized transcriptomics data from healthy renal tubular cells exposed to healthy and post-dialysis uremic plasma. RESULTS Differential gene expression analysis identified 983 up-regulated genes, including 70 essential proteins in the protein-protein interaction network. Modularity-based clustering revealed six clusters of essential proteins associated with 11 pathological pathways activated in response to non-dialyzable uremic toxins. CONCLUSIONS Notably, WNT1/11, AGT, FGF4/17/22, LMX1B, GATA4, and CXCL12 emerged as promising targets for further exploration in renal tubular pathology related to non-dialyzable uremic toxins. Understanding the molecular players and pathways linked to renal tubular dysfunction opens avenues for novel therapeutic interventions and improved clinical management of chronic kidney disease and its complications.
Collapse
Affiliation(s)
- Roya Asadi
- Industrial Engineering Department, Faculty of Technical and Engineering, University of Science and Culture (USC), Tehran, Iran
| | - Pejman Shadpour
- Hospital Management Research Center (HMRC), Hasheminejad Kidney Center (HKC), Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Akram Nakhaei
- Computer Engineering Department, Mazandaran University of Science and Technology (MUST), Babol, Iran.
| |
Collapse
|
2
|
Shah SN, Knausenberger TBA, Pontifex MG, Connell E, Le Gall G, Hardy TA, Randall DW, McCafferty K, Yaqoob MM, Solito E, Müller M, Stachulski AV, Glen RC, Vauzour D, Hoyles L, McArthur S. Cerebrovascular damage caused by the gut microbe/host co-metabolite p-cresol sulfate is prevented by blockade of the EGF receptor. Gut Microbes 2024; 16:2431651. [PMID: 39582109 PMCID: PMC11591591 DOI: 10.1080/19490976.2024.2431651] [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: 08/07/2024] [Revised: 10/15/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
The gut microbiota-brain axis has been associated with the pathogenesis of numerous disorders, but the mechanism(s) underlying these links are generally poorly understood. Accumulating evidence indicates the involvement of gut microbe-derived metabolites. Circulating levels of the gut microbe/host co-metabolite p-cresol sulfate (pCS) correlate with cerebrovascular event risk in individuals with chronic kidney disease (CKD), but whether this relationship is mechanistic is unclear. We hypothesized that pCS would impair the function of the blood-brain barrier (BBB), the primary brain vasculature interface. We report that pCS exposure impairs BBB integrity in human cells in vitro and both acutely (≤6 hours) and chronically (28 days) in mice, enhancing tracer extravasation, disrupting barrier-regulating tight junction components and ultimately exerting a suppressive effect upon whole-brain transcriptomic activity. In vitro and in vivo mechanistic studies showed that pCS activated epidermal growth factor receptor (EGFR) signaling, sequentially activating the intracellular signaling proteins annexin A1 and STAT3 to induce mobilization of matrix metalloproteinase MMP-2/9 and disruption to the integrity of the BBB. This effect was confirmed as specific to the EGFR through the use of both pharmacological and RNA interference approaches. Confirming the translational relevance of this work, exposure of the cerebromicrovascular endothelia to serum from hemodialysis patients in vitro led to a significant increase in paracellular permeability, with the magnitude of permeabilization closely correlating with serum pCS, but not most other uremic toxin, content. Notably, this damaging effect of hemodialysis patient serum was prevented by pharmacological blockade of the EGFR. Our results define a pathway linking the co-metabolite pCS with BBB damage and suggest that targeting the EGFR may mitigate against cerebrovascular damage in CKD. This work further provides mechanistic evidence indicating the role of gut microbe-derived metabolites in human disease.
Collapse
Affiliation(s)
- Sita N. Shah
- Blizard Institute, Faculty of Medicine & Dentistry, Queen Mary, University of London, London, UK
| | | | - Matthew G. Pontifex
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich, UK
| | - Emily Connell
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich, UK
| | - Gwénaëlle Le Gall
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich, UK
| | - Tom A.J. Hardy
- Blizard Institute, Faculty of Medicine & Dentistry, Queen Mary, University of London, London, UK
| | - David W. Randall
- Department of Renal Medicine and Transplantation, Royal London Hospital, Barts Health NHS Trust, London, UK
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary, University of London, London, UK
| | - Kieran McCafferty
- Department of Renal Medicine and Transplantation, Royal London Hospital, Barts Health NHS Trust, London, UK
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary, University of London, London, UK
| | - Muhammad M. Yaqoob
- Department of Renal Medicine and Transplantation, Royal London Hospital, Barts Health NHS Trust, London, UK
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary, University of London, London, UK
| | - Egle Solito
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary, University of London, London, UK
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Michael Müller
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich, UK
| | - Andrew V. Stachulski
- Robert Robinson Laboratories, Department of Chemistry, University of Liverpool, Liverpool, UK
| | - Robert C. Glen
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - David Vauzour
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich, UK
| | - Lesley Hoyles
- Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, UK
| | - Simon McArthur
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary, University of London, London, UK
| |
Collapse
|
3
|
Yang Y, Mihajlovic M, Masereeuw R. Protein-Bound Uremic Toxins in Senescence and Kidney Fibrosis. Biomedicines 2023; 11:2408. [PMID: 37760849 PMCID: PMC10525416 DOI: 10.3390/biomedicines11092408] [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: 08/07/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition of kidney dysfunction due to diverse causes of injury. In healthy kidneys, protein-bound uremic toxins (PBUTs) are cleared from the systemic circulation by proximal tubule cells through the concerted action of plasma membrane transporters that facilitate their urinary excretion, but the endogenous metabolites are hardly removed with kidney dysfunction and may contribute to CKD progression. Accumulating evidence suggests that senescence of kidney tubule cells influences kidney fibrosis, the common endpoint for CKD with an excessive accumulation of extracellular matrix (ECM). Senescence is a special state of cells characterized by permanent cell cycle arrest and limitation of proliferation, which promotes fibrosis by releasing senescence-associated secretory phenotype (SASP) factors. The accumulation of PBUTs in CKD causes oxidative stress and increases the production of inflammatory (SASP) factors that could trigger fibrosis. Recent studies gave some clues that PBUTs may also promote senescence in kidney tubular cells. This review provides an overview on how senescence contributes to CKD, the involvement of PBUTs in this process, and how kidney senescence can be studied. Finally, some suggestions for future therapeutic options for CKD while targeting senescence are given.
Collapse
Affiliation(s)
- Yi Yang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Milos Mihajlovic
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| |
Collapse
|
4
|
Zhang RB, Dong LC, Huang Q, Shen Y, Li HY, Yu SG, Wu QF. Matrix metalloproteinases are key targets of acupuncture in the treatment of ulcerative colitis. Exp Biol Med (Maywood) 2023; 248:1229-1241. [PMID: 37438919 PMCID: PMC10621479 DOI: 10.1177/15353702231182205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 04/10/2023] [Indexed: 07/14/2023] Open
Abstract
The aim of this study was to elucidate the key targets of acupuncture in the colon of ulcerative colitis (UC) mice model using full-length transcriptome sequencing. 2.5% dextran sodium sulfate (DSS)-induced colitis mice were treated with or without acupuncture. Intestinal pathology was observed, and full transcriptome sequencing and bioinformatic analysis were performed. The results demonstrated that acupuncture treatment reduced the UC symptoms, disease activity index score, and histological colitis score and increased body weight, colon length, and the number of intestinal goblet cells. In addition, acupuncture can also decrease the expression of necrotic biomarker phosphorylates mixed lineage kinase domain-like pseudo kinase (p-MLKL). Full-length transcriptome analysis indicated that acupuncture reversed the expression of 987 of the 1918 upregulated differentially expressed genes (DEGs), and 632 of the 1351 downregulated DEGs induced by DSS. DEGs regulated by acupuncture were mainly involved in inflammatory responses and intestinal barrier pathways. The protein-protein interaction network analysis revealed that matrix metalloproteinases (MMPs) are important genes regulated by acupuncture. Gene set enrichment analysis revealed that extracellular matrix (ECM)-receptor interaction was an important target of acupuncture. In addition, alternative splicing analysis suggested that acupuncture improved signaling pathways related to intestinal permeability, the biological processes of xenobiotics, sulfur compounds, and that monocarboxylic acids are closely associated with MMPs. Overall, our transcriptome analysis results indicate that acupuncture improves intestinal barrier function in UC through negative regulation of MMPs expression.
Collapse
Affiliation(s)
| | | | - Qin Huang
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yuan Shen
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Hong-Ying Li
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shu-Guang Yu
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Qiao-Feng Wu
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
5
|
Abstract
Homeostasis is a prerequisite for health. When homeostasis becomes disrupted, dysfunction occurs. This is especially the case for the gut microbiota, which under normal conditions lives in symbiosis with the host. As there are as many microbial cells in and on our body as human cells, it is unlikely they would not contribute to health or disease. The gut bacterial metabolism generates numerous beneficial metabolites but also uremic toxins and their precursors, which are transported into the circulation. Barrier function in the intestine, the heart, and the kidneys regulates metabolite transport and concentration and plays a role in inter-organ and inter-organism communication via small molecules. This communication is analyzed from the perspective of the remote sensing and signaling theory, which emphasizes the role of a large network of multispecific, oligospecific, and monospecific transporters and enzymes in regulating small-molecule homeostasis. The theory provides a systems biology framework for understanding organ cross talk and microbe-host communication involving metabolites, signaling molecules, nutrients, antioxidants, and uremic toxins. This remote small-molecule communication is critical for maintenance of homeostasis along the gut-heart-kidney axis and for responding to homeostatic perturbations. Chronic kidney disease is characterized by gut dysbiosis and accumulation of toxic metabolites. This slowly impacts the body, affecting the cardiovascular system and contributing to the progression of kidney dysfunction, which in its turn influences the gut microbiota. Preserving gut homeostasis and barrier functions or restoring gut dysbiosis and dysfunction could be a minimally invasive way to improve patient outcomes and quality of life in many diseases, including cardiovascular and kidney disease.
Collapse
Affiliation(s)
- Griet Glorieux
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| | - Sanjay K Nigam
- Department of Pediatrics (S.K.N.), University of California San Diego, La Jolla, CA
- Division of Nephrology, Department of Medicine (S.K.N.), University of California San Diego, La Jolla, CA
| | - Raymond Vanholder
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| | - Francis Verbeke
- Nephrology Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Gent, Belgium (G.G., R.V., F.V.)
| |
Collapse
|
6
|
Liabeuf S, Drueke T, Massy Z. Rôle des toxines urémiques dans la genèse des complications de la maladie rénale chronique. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2023. [DOI: 10.1016/j.banm.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
7
|
The Evolving View of Uremic Toxicity. Toxins (Basel) 2022; 14:toxins14040274. [PMID: 35448883 PMCID: PMC9031373 DOI: 10.3390/toxins14040274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
Indoxyl sulfate, closely related to indigo, a dye valued for it binding to cloth, has been recognized as a protein-bound solute bound to albumin, present in increased concentration in the serum of patients with impaired glomerular filtration (13). The early studies of Niwa identified indoxyl sulfate as a toxin capable of accelerating the rate of renal damage in subtotal nephrectomized rats (18). Over the past decade other protein-bound solutes have been identified in the plasma of patients with impaired glomerular filtration. Although the early studies, focused on the kidney, identified indoxyl sulfate as a toxic waste product dependent on the kidney for its removal, subsequent observations have identified organic anion transporters on many non-renal tissue, leading to the view that indoxyl sulfate is part of a systemic signaling system.
Collapse
|
8
|
Mihajlovic M, Krebber MM, Yang Y, Ahmed S, Lozovanu V, Andreeva D, Verhaar MC, Masereeuw R. Protein-Bound Uremic Toxins Induce Reactive Oxygen Species-Dependent and Inflammasome-Mediated IL-1β Production in Kidney Proximal Tubule Cells. Biomedicines 2021; 9:biomedicines9101326. [PMID: 34680443 PMCID: PMC8533138 DOI: 10.3390/biomedicines9101326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/19/2021] [Accepted: 09/23/2021] [Indexed: 12/29/2022] Open
Abstract
Protein bound-uremic toxins (PBUTs) are not efficiently removed by hemodialysis in chronic kidney disease (CKD) patients and their accumulation leads to various co-morbidities via cellular dysfunction, inflammation and oxidative stress. Moreover, it has been shown that increased intrarenal expression of the NLRP3 receptor and IL-1β are associated with reduced kidney function, suggesting a critical role for the NLRP3 inflammasome in CKD progression. Here, we evaluated the effect of PBUTs on inflammasome-mediated IL-1β production in vitro and in vivo. Exposure of human conditionally immortalized proximal tubule epithelial cells to indoxyl sulfate (IS) and a mixture of anionic PBUTs (UT mix) increased expression levels of NLRP3, caspase-1 and IL-1β, accompanied by a significant increase in IL-1β secretion and caspase-1 activity. Furthermore, IS and UT mix induced the production of intracellular reactive oxygen species, and caspase-1 activity and IL-1β secretion were reduced in the presence of antioxidant N-acetylcysteine. IS and UT mix also induced NF-κB activation as evidenced by p65 nuclear translocation and IL-1β production, which was counteracted by an IKK inhibitor. In vivo, using subtotal nephrectomy CKD rats, a significant increase in total plasma levels of IS and the PBUTs, kynurenic acid and hippuric acid, was found, as well as enhanced urinary malondialdehyde levels. CKD kidney tissue showed an increasing trend in expression of NLRP3 inflammasome components, and a decreasing trend in superoxide dismutase-1 levels. In conclusion, we showed that PBUTs induce inflammasome-mediated IL-1β production in proximal tubule cells via oxidative stress and NF-κB signaling, suggesting their involvement in disease-associated inflammatory processes.
Collapse
Affiliation(s)
- Milos Mihajlovic
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.M.); (Y.Y.); (S.A.); (V.L.); (D.A.)
| | - Merle M. Krebber
- Department Nephrology and Hypertension, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (M.M.K.); (M.C.V.)
| | - Yi Yang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.M.); (Y.Y.); (S.A.); (V.L.); (D.A.)
| | - Sabbir Ahmed
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.M.); (Y.Y.); (S.A.); (V.L.); (D.A.)
| | - Valeria Lozovanu
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.M.); (Y.Y.); (S.A.); (V.L.); (D.A.)
| | - Daria Andreeva
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.M.); (Y.Y.); (S.A.); (V.L.); (D.A.)
| | - Marianne C. Verhaar
- Department Nephrology and Hypertension, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (M.M.K.); (M.C.V.)
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.M.); (Y.Y.); (S.A.); (V.L.); (D.A.)
- Correspondence:
| |
Collapse
|
9
|
Rodriguez-Coira J, Villaseñor A, Izquierdo E, Huang M, Barker-Tejeda TC, Radzikowska U, Sokolowska M, Barber D. The Importance of Metabolism for Immune Homeostasis in Allergic Diseases. Front Immunol 2021; 12:692004. [PMID: 34394086 PMCID: PMC8355700 DOI: 10.3389/fimmu.2021.692004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022] Open
Abstract
There is increasing evidence that the metabolic status of T cells and macrophages is associated with severe phenotypes of chronic inflammation, including allergic inflammation. Metabolic changes in immune cells have a crucial role in their inflammatory or regulatory responses. This notion is reinforced by metabolic diseases influencing global energy metabolism, such as diabetes or obesity, which are known risk factors of severity in inflammatory conditions, due to the metabolic-associated inflammation present in these patients. Since several metabolic pathways are closely tied to T cell and macrophage differentiation, a better understanding of metabolic alterations in immune disorders could help to restore and modulate immune cell functions. This link between energy metabolism and inflammation can be studied employing animal, human or cellular models. Analytical approaches rank from classic immunological studies to integrated analysis of metabolomics, transcriptomics, and proteomics. This review summarizes the main metabolic pathways of the cells involved in the allergic reaction with a focus on T cells and macrophages and describes different models and platforms of analysis used to study the immune system and its relationship with metabolism.
Collapse
Affiliation(s)
- Juan Rodriguez-Coira
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Alma Villaseñor
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Elena Izquierdo
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Tomás Clive Barker-Tejeda
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Domingo Barber
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| |
Collapse
|
10
|
Young GH, Lin JT, Cheng YF, Ho CF, Kuok QY, Hsu RC, Liao WR, Chen CC, Chen HM. Modulation of adenine phosphoribosyltransferase-mediated salvage pathway to accelerate diabetic wound healing. FASEB J 2021; 35:e21296. [PMID: 33675115 DOI: 10.1096/fj.202001736rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 01/13/2023]
Abstract
Adenine phosphoribosyltransferase (APRT) is the key enzyme involved in purine salvage by the incorporation of adenine and phosphoribosyl pyrophosphate to provide adenylate nucleotides. To evaluate the role of APRT in the repair processes of cutaneous wounds in healthy skin and in diabetic patients, a diabetic mouse model (db/db) and age-matched wild-type mice were used. Moreover, the topical application of adenine was assessed. In vitro studies, analytical, histological, and immunohistochemical methods were used. Diabetic mice treated with adenine exhibited elevated ATP levels in organismic skin and accelerated wound healing. In vitro studies showed that APRT utilized adenine to rescue cellular ATP levels and proliferation from hydrogen peroxide-induced oxidative damage. HPLC-ESI-MS/MS-based analysis of total adenylate nucleotides in NIH-3T3 fibroblasts demonstrated that adenine addition enlarged the cellular adenylate pool, reduced the adenylate energy charge, and provided additional AMP for the further generation of ATP. These data indicate an upregulation of APRT in skin wounds, highlighting its role during the healing of diabetic wounds through regulation of the nucleotide pool after injury. Furthermore, topical adenine supplementation resulted in an enlargement of the adenylate pool needed for the generation of ATP, an important molecule for wound repair.
Collapse
Affiliation(s)
| | | | | | | | | | - Ru-Chun Hsu
- Energenesis Biomedical Co. Ltd, Taipei, Taiwan
| | | | | | - Han-Min Chen
- Energenesis Biomedical Co. Ltd, Taipei, Taiwan.,Department of Life Science, Institute of Applied Science and Engineering, Catholic Fu-Jen University, New Taipei City, Taiwan
| |
Collapse
|
11
|
Pomyen Y, Budhu A, Chaisaingmongkol J, Forgues M, Dang H, Ruchirawat M, Mahidol C, Wang XW. Tumor metabolism and associated serum metabolites define prognostic subtypes of Asian hepatocellular carcinoma. Sci Rep 2021; 11:12097. [PMID: 34103600 PMCID: PMC8187378 DOI: 10.1038/s41598-021-91560-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/24/2021] [Indexed: 12/20/2022] Open
Abstract
Treatment effectiveness in hepatocellular carcinoma (HCC) depends on early detection and precision-medicine-based patient stratification for targeted therapies. However, the lack of robust biomarkers, particularly a non-invasive diagnostic tool, precludes significant improvement of clinical outcomes for HCC patients. Serum metabolites are one of the best non-invasive means for determining patient prognosis, as they are stable end-products of biochemical processes in human body. In this study, we aimed to identify prognostic serum metabolites in HCC. To determine serum metabolites that were relevant and representative of the tissue status, we performed a two-step correlation analysis to first determine associations between metabolic genes and tissue metabolites, and second, between tissue metabolites and serum metabolites among 49 HCC patients, which were then validated in 408 additional Asian HCC patients with mixed etiologies. We found that certain metabolic genes, tissue metabolites and serum metabolites can independently stratify HCC patients into prognostic subgroups, which are consistent across these different data types and our previous findings. The metabolic subtypes are associated with β-oxidation process in fatty acid metabolism, where patients with worse survival outcome have dysregulated fatty acid metabolism. These serum metabolites may be used as non-invasive biomarkers to define prognostic tumor molecular subtypes for HCC.
Collapse
Affiliation(s)
- Yotsawat Pomyen
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.,Translational Research Unit, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Anuradha Budhu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.,Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Jittiporn Chaisaingmongkol
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.,Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand.,Center of Excellence on Environmental Health and Toxicology, Office of Higher Education Commission, Ministry of Higher Education, Science, Research and Innovation, Bangkok, 10400, Thailand
| | - Marshonna Forgues
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Hien Dang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.,Division of Surgery, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mathuros Ruchirawat
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand.,Center of Excellence on Environmental Health and Toxicology, Office of Higher Education Commission, Ministry of Higher Education, Science, Research and Innovation, Bangkok, 10400, Thailand
| | - Chulabhorn Mahidol
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA. .,Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.
| | | |
Collapse
|
12
|
Lowenstein J, Nigam SK. Uremic Toxins in Organ Crosstalk. Front Med (Lausanne) 2021; 8:592602. [PMID: 33937275 PMCID: PMC8085272 DOI: 10.3389/fmed.2021.592602] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 03/18/2021] [Indexed: 11/13/2022] Open
Abstract
Many putative uremic toxins—like indoxyl sulfate, p-cresol sulfate, kynurenic acid, uric acid, and CMPF—are organic anions. Both inter-organ and inter-organismal communication are involved. For example, the gut microbiome is the main source of indole, which, after modification by liver drug metabolizing enzymes (DMEs), becomes indoxyl sulfate. Various organic anion transporters (organic anion transporters, OATs; organic anion-transporting polypeptides, OATPs; multidrug resistance-associated proteins, MRPs, and other ABC transporters like ABCG2)—often termed “drug transporters”—mediate movement of uremic toxins through cells and organs. In the kidney proximal tubule, critical roles for OAT1 and OAT3 in regulating levels of protein-bound uremic toxins have been established using knock-out mice. OATs are important in maintaining residual tubular function in chronic kidney disease (CKD); as CKD progresses, intestinal transporters like ABCG2, which extrude urate and other organic anions into the gut lumen, seem to help restore homeostasis. Uremic toxins like indoxyl sulfate also regulate signaling and metabolism, potentially affecting gene expression in extra-renal tissues as well as the kidney. Focusing on the history and evolving story of indoxyl sulfate, we discuss how uremic toxins appear to be part of an extensive “remote sensing and signaling” network—involving so-called drug transporters and drug metabolizing enzymes which modulate metabolism and signaling. This systems biology view of uremic toxins is leading to a new appreciation of uremia as partly due to disordered remote sensing and signaling mechanisms–resulting from, and causing, aberrant inter-organ (e.g., gut-liver- kidney-CNS) and inter-organismal (e.g., gut microbiome-host) communication.
Collapse
Affiliation(s)
- Jerome Lowenstein
- Department of Nephrology, New York University School of Medicine, New York, NY, United States
| | - Sanjay K Nigam
- Departments of Pediatrics and Medicine (Nephrology), San Diego School of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
13
|
Favero C, Carriazo S, Cuarental L, Fernandez-Prado R, Gomá-Garcés E, Perez-Gomez MV, Ortiz A, Fernandez-Fernandez B, Sanchez-Niño MD. Phosphate, Microbiota and CKD. Nutrients 2021; 13:1273. [PMID: 33924419 PMCID: PMC8070653 DOI: 10.3390/nu13041273] [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: 03/07/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 02/08/2023] Open
Abstract
Phosphate is a key uremic toxin associated with adverse outcomes. As chronic kidney disease (CKD) progresses, the kidney capacity to excrete excess dietary phosphate decreases, triggering compensatory endocrine responses that drive CKD-mineral and bone disorder (CKD-MBD). Eventually, hyperphosphatemia develops, and low phosphate diet and phosphate binders are prescribed. Recent data have identified a potential role of the gut microbiota in mineral bone disorders. Thus, parathyroid hormone (PTH) only caused bone loss in mice whose microbiota was enriched in the Th17 cell-inducing taxa segmented filamentous bacteria. Furthermore, the microbiota was required for PTH to stimulate bone formation and increase bone mass, and this was dependent on bacterial production of the short-chain fatty acid butyrate. We review current knowledge on the relationship between phosphate, microbiota and CKD-MBD. Topics include microbial bioactive compounds of special interest in CKD, the impact of dietary phosphate and phosphate binders on the gut microbiota, the modulation of CKD-MBD by the microbiota and the potential therapeutic use of microbiota to treat CKD-MBD through the clinical translation of concepts from other fields of science such as the optimization of phosphorus utilization and the use of phosphate-accumulating organisms.
Collapse
Affiliation(s)
- Chiara Favero
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
| | - Sol Carriazo
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
- Red de Investigacion Renal (REDINREN), Av Reyes Católicos 2, 28040 Madrid, Spain
| | - Leticia Cuarental
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
- Red de Investigacion Renal (REDINREN), Av Reyes Católicos 2, 28040 Madrid, Spain
| | - Raul Fernandez-Prado
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
- Red de Investigacion Renal (REDINREN), Av Reyes Católicos 2, 28040 Madrid, Spain
| | - Elena Gomá-Garcés
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
| | - Maria Vanessa Perez-Gomez
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
- Red de Investigacion Renal (REDINREN), Av Reyes Católicos 2, 28040 Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
- Red de Investigacion Renal (REDINREN), Av Reyes Católicos 2, 28040 Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
- Red de Investigacion Renal (REDINREN), Av Reyes Católicos 2, 28040 Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Av Reyes Católicos 2, 28040 Madrid, Spain; (C.F.); (S.C.); (L.C.); (R.F.-P.); (E.G.-G.); (M.V.P.-G.)
- Red de Investigacion Renal (REDINREN), Av Reyes Católicos 2, 28040 Madrid, Spain
- School of Medicine, Department of Pharmacology and Therapeutics, Universidad Autonoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
14
|
JNK and p38 Inhibitors Prevent Transforming Growth Factor-β1-Induced Myofibroblast Transdifferentiation in Human Graves' Orbital Fibroblasts. Int J Mol Sci 2021; 22:ijms22062952. [PMID: 33799469 PMCID: PMC7998969 DOI: 10.3390/ijms22062952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1)-induced myofibroblast transdifferentiation from orbital fibroblasts is known to dominate tissue remodeling and fibrosis in Graves’ ophthalmopathy (GO). However, the signaling pathways through which TGF-β1 activates Graves’ orbital fibroblasts remain unclear. This study investigated the role of the mitogen-activated protein kinase (MAPK) pathway in TGF-β1-induced myofibroblast transdifferentiation in human Graves’ orbital fibroblasts. The MAPK pathway was assessed by measuring the phosphorylation of p38, c-Jun N-terminal kinase (JNK), and extracellular-signal-regulated kinase (ERK) by Western blots. The expression of connective tissue growth factor (CTGF), α-smooth muscle actin (α-SMA), and fibronectin representing fibrogenesis was estimated. The activities of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) responsible for extracellular matrix (ECM) metabolism were analyzed. Specific pharmacologic kinase inhibitors were used to confirm the involvement of the MAPK pathway. After treatment with TGF-β1, the phosphorylation levels of p38 and JNK, but not ERK, were increased. CTGF, α-SMA, and fibronectin, as well as TIMP-1 and TIMP-3, were upregulated, whereas the activities of MMP-2/-9 were inhibited. The effects of TGF-β1 on the expression of these factors were eliminated by p38 and JNK inhibitors. The results suggested that TGF-β1 could induce myofibroblast transdifferentiation in human Graves’ orbital fibroblasts through the p38 and JNK pathways.
Collapse
|
15
|
Wypych TP, Pattaroni C, Perdijk O, Yap C, Trompette A, Anderson D, Creek DJ, Harris NL, Marsland BJ. Microbial metabolism of L-tyrosine protects against allergic airway inflammation. Nat Immunol 2021; 22:279-286. [PMID: 33495652 DOI: 10.1038/s41590-020-00856-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/03/2020] [Indexed: 01/29/2023]
Abstract
The constituents of the gut microbiome are determined by the local habitat, which itself is shaped by immunological pressures, such as mucosal IgA. Using a mouse model of restricted antibody repertoire, we identified a role for antibody-microbe interactions in shaping a community of bacteria with an enhanced capacity to metabolize L-tyrosine. This model led to increased concentrations of p-cresol sulfate (PCS), which protected the host against allergic airway inflammation. PCS selectively reduced CCL20 production by airway epithelial cells due to an uncoupling of epidermal growth factor receptor (EGFR) and Toll-like receptor 4 (TLR4) signaling. Together, these data reveal a gut microbe-derived metabolite pathway that acts distally on the airway epithelium to reduce allergic airway responses, such as those underpinning asthma.
Collapse
Affiliation(s)
- Tomasz P Wypych
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Céline Pattaroni
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Olaf Perdijk
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Carmen Yap
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Aurélien Trompette
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Epalinges, Switzerland
| | - Dovile Anderson
- Monash Proteomics and Metabolomics Facility, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Darren J Creek
- Monash Proteomics and Metabolomics Facility, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
16
|
O’Brien FJ, Mair RD, Plummer NS, Meyer TW, Sutherland SM, Sirich TL. Impaired Tubular Secretion of Organic Solutes in Acute Kidney Injury. KIDNEY360 2020; 1:724-730. [PMID: 35252876 PMCID: PMC8815732 DOI: 10.34067/kid.0001632020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/24/2020] [Indexed: 06/14/2023]
Abstract
BACKGROUND Impairment of kidney function is routinely assessed by measuring the accumulation of creatinine, an organic solute cleared largely by glomerular filtration. We tested whether the clearance of solutes that undergo tubular secretion is reduced in proportion to the clearance of creatinine in humans with AKI. METHODS Four endogenously produced organic solutes (phenylacetylglutamine [PAG], hippurate [HIPP], indoxyl sulfate [IS], and p-cresol sulfate [PCS]) were measured in spot urine and plasma samples from ten patients with AKI and 17 controls. Fractional clearance relative to creatinine was calculated to assess tubular secretion. Fractional clearance values were calculated in terms of the free, unbound levels of HIPP, IS, and PCS that bind to plasma proteins. RESULTS Fractional clearance values for PAG, HIPP, IS, and PCS were >1.0 in patients with AKI as well as controls, indicating that these solutes were still secreted by the tubules of the injured kidneys. Fractional clearance values were, however, significantly lower in patients with AKI than controls, indicating that kidney injury reduced tubular secretion more than glomerular filtration (AKI versus control: PAG, 2.1±0.7 versus 4.6±1.4, P<0.001; HIPP, 10±5 versus 15±7, P=0.02; IS, 10±6 versus 28±7, P<0.001; PCS, 3.3±1.8 versus 10±3, P<0.001). Free plasma levels rose out of proportion to total plasma levels for each of the bound solutes in AKI, so that calculating their fractional clearance in terms of their total plasma levels failed to reveal their impaired secretion. CONCLUSIONS Tubular secretion of organic solutes can be reduced out of proportion to glomerular filtration in AKI. Impaired secretion of protein-bound solutes may be more reliably detected when clearances are expressed in terms of their free, unbound levels in the plasma.
Collapse
Affiliation(s)
- Frank J. O’Brien
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Robert D. Mair
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Natalie S. Plummer
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Timothy W. Meyer
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Scott M. Sutherland
- Department of Pediatrics, Lucile Packard Children’s Hospital, Stanford University, Palo Alto, California
| | - Tammy L. Sirich
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
17
|
Sun B, Wang X, Liu X, Wang L, Ren F, Wang X, Leng X. Hippuric Acid Promotes Renal Fibrosis by Disrupting Redox Homeostasis via Facilitation of NRF2-KEAP1-CUL3 Interactions in Chronic Kidney Disease. Antioxidants (Basel) 2020; 9:antiox9090783. [PMID: 32854194 PMCID: PMC7555723 DOI: 10.3390/antiox9090783] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/16/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by the accumulation of protein-bound uremic toxins (PBUTs), which play a pathophysiological role in renal fibrosis (a common pathological process resulting in CKD progression). Accumulation of the PBUT hippuric acid (HA) is positively correlated with disease progression in CKD patients, suggesting that HA may promote renal fibrosis. Oxidative stress is the most important factor affecting PBUTs nephrotoxicity. Herein, we assessed the ability of HA to promote kidney fibrosis by disrupting redox homeostasis. In HK-2 cells, HA increased fibrosis-related gene expression, extracellular matrix imbalance, and oxidative stress. Additionally, reactive oxygen species (ROS)-mediated TGFβ/SMAD signaling contributed to HA-induced fibrotic responses. HA disrupted antioxidant networks by decreasing the levels of nuclear factor erythroid 2-related factor 2 (NRF2), leading to ROS accumulation and fibrotic responses, as evidenced by NRF2 activation and knockdown. Moreover, NRF2 levels were reduced by NRF2 ubiquitination, which was regulated via increased interactions of Kelch-like ECH-associated protein 1 with Cullin 3 and NRF2. Finally, renal fibrosis and redox imbalance promoted by HA were confirmed in rats. Importantly, sulforaphane (NRF2 activator) reversed HA-promoted renal fibrosis. Thus, HA promotes renal fibrosis in CKD by disrupting NRF2-driven antioxidant system, indicating that NRF2 is a potential therapeutic target for CKD.
Collapse
Affiliation(s)
- Bowen Sun
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.S.); (X.W.); (X.L.); (L.W.); (F.R.)
| | - Xifan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.S.); (X.W.); (X.L.); (L.W.); (F.R.)
| | - Xiaoxue Liu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.S.); (X.W.); (X.L.); (L.W.); (F.R.)
| | - Longjiao Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.S.); (X.W.); (X.L.); (L.W.); (F.R.)
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.S.); (X.W.); (X.L.); (L.W.); (F.R.)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xiaoyu Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.S.); (X.W.); (X.L.); (L.W.); (F.R.)
- Correspondence: (X.W.); (X.L.); Tel.: +86-10-6273-8589 (X.W.); +86-10-6273-7761 (X.L.)
| | - Xiaojing Leng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.S.); (X.W.); (X.L.); (L.W.); (F.R.)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Correspondence: (X.W.); (X.L.); Tel.: +86-10-6273-8589 (X.W.); +86-10-6273-7761 (X.L.)
| |
Collapse
|
18
|
How do Uremic Toxins Affect the Endothelium? Toxins (Basel) 2020; 12:toxins12060412. [PMID: 32575762 PMCID: PMC7354502 DOI: 10.3390/toxins12060412] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
Uremic toxins can induce endothelial dysfunction in patients with chronic kidney disease (CKD). Indeed, the structure of the endothelial monolayer is damaged in CKD, and studies have shown that the uremic toxins contribute to the loss of cell–cell junctions, increasing permeability. Membrane proteins, such as transporters and receptors, can mediate the interaction between uremic toxins and endothelial cells. In these cells, uremic toxins induce oxidative stress and activation of signaling pathways, including the aryl hydrocarbon receptor (AhR), nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) pathways. The activation of these pathways leads to overexpression of proinflammatory (e.g., monocyte chemoattractant protein-1, E-selectin) and prothrombotic (e.g., tissue factor) proteins. Uremic toxins also induce the formation of endothelial microparticles (EMPs), which can lead to the activation and dysfunction of other cells, and modulate the expression of microRNAs that have an important role in the regulation of cellular processes. The resulting endothelial dysfunction contributes to the pathogenesis of cardiovascular diseases, such as atherosclerosis and thrombotic events. Therefore, uremic toxins as well as the pathways they modulated may be potential targets for therapies in order to improve treatment for patients with CKD.
Collapse
|
19
|
Young GH, Tang SC, Wu VC, Wang KC, Nong JY, Huang PY, Hu CJ, Chiou HY, Jeng JS, Hsu CY. The functional role of hemojuvelin in acute ischemic stroke. J Cereb Blood Flow Metab 2020; 40:1316-1327. [PMID: 31307288 PMCID: PMC7238368 DOI: 10.1177/0271678x19861448] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Our study aimed to establish the role of hemojuvelin (HJV) in acute ischemic stroke (AIS). We performed immunohistochemistry for HJV expression in human brain tissues from 10 AIS and 2 non-stroke autopsy subjects. Plasma HJV was measured in 112 AIS patients within 48 h after stroke. The results showed significantly increased HJV expression in brain tissues from AIS patients compare to non-stroke subjects. After adjusting for clinical variables, plasma levels of HJV within 48 h after stroke were an independent predictor of poor functional outcome three months post-stroke (OR:1.78, 95% CI: 1.03-3.07; P = 0.038). In basic part, Western blotting showed that HJV expression in mice brains was apparent at 3 h after middle cerebral artery occlusion (MCAO), and increased significantly at 72 h. In cultured cortical neurons, expression of HJV protein increased remarkably 24 h after oxygen glucose deprivation (OGD), and small interfering RNAs (siHJV) transfected OGD neurons had a lower apoptotic rate. Importantly, 72 h post-MCAO, HJV knockout mice had significantly smaller infarcts and less expression of cleaved caspase-3 protein compared with wild-type mice. In summary, HJV participates in the mechanisms of post-stroke neuronal injury, and that plasma HJV levels can be a potential early outcome indicator for AIS patients.
Collapse
Affiliation(s)
| | - Sung-Chun Tang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei
| | - Vin-Cent Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Kuo-Chuan Wang
- Department of Surgery, National Taiwan University Hospital, Taipei
| | - Jing-Yi Nong
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Po-Yuan Huang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei
| | - Chaur-Jong Hu
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, Taipei
| | - Hung-Yi Chiou
- School of Public Health, Taipei Medical University, Taipei
| | - Jiann-Shing Jeng
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei
| | - Chung Y Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung
| |
Collapse
|
20
|
Oral Charcoal Adsorbents Attenuate Neointima Formation of Arteriovenous Fistulas. Toxins (Basel) 2020; 12:toxins12040237. [PMID: 32276394 PMCID: PMC7232464 DOI: 10.3390/toxins12040237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/31/2020] [Accepted: 04/05/2020] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) accelerates the development of neointima formation at the anastomosis site of arteriovenous (AV) fistulas. Accumulation of certain uremic toxins has a deleterious effect on the cardiovascular system. The oral charcoal adsorbent, AST-120, reduces circulating and tissue uremic toxins, but its effect on neointima formation at an AV fistula is unknown. To understand the effect of CKD and AST-120 on neointima formation, we created AV fistulas (common carotid artery to the external jugular vein in an end-to-side anastomosis) in mice with and without CKD. AST-120 was administered in chow before and after AV fistula creation. Administration of AST-120 significantly decreased serum indoxyl sulfate levels in CKD mice. CKD mice had a larger neointima area than non-CKD mice, and administration of AST-120 in CKD mice attenuated neointima formation. Both smooth muscle cell and fibrin components were increased in CKD mice, and AST-120 decreased both. RNA expression of MMP-2, MMP-9, TNFα, and TGFβ was increased in neointima tissue of CKD mice, and AST-120 administration neutralized the expression. Our results provided in vivo evidence to support the role of uremic toxin-binding therapy on the prevention of neointima formation. Peri-operative AST-120 administration deserves further investigation as a potential therapy to improve AV fistula patency.
Collapse
|
21
|
Al-Jaishi AA, McIntyre CW, Sontrop JM, Dixon SN, Anderson S, Bagga A, Benjamin D, Berry D, Blake PG, Chambers L, Chan PCK, Delbrouck N, Devereaux PJ, Ferreira-Divino LF, Goluch R, Gregor L, Grimshaw JM, Hanson G, Iliescu E, Jain AK, Lok CE, Mustafa RA, Nathoo B, Nesrallah GE, Oliver MJ, Pandeya S, Parmar MS, Perkins D, Presseau J, Rabin E, Sasal J, Shulman T, Sood MM, Steele A, Tam P, Tascona D, Wadehra D, Wald R, Walsh M, Watson P, Wodchis W, Zager P, Zwarenstein M, Garg AX. Major Outcomes With Personalized Dialysate TEMPerature (MyTEMP): Rationale and Design of a Pragmatic, Registry-Based, Cluster Randomized Controlled Trial. Can J Kidney Health Dis 2020; 7:2054358119887988. [PMID: 32076569 PMCID: PMC7003172 DOI: 10.1177/2054358119887988] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/23/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Small randomized trials demonstrated that a lower compared with higher dialysate temperature reduced the average drop in intradialytic blood pressure. Some observational studies demonstrated that a lower compared with higher dialysate temperature was associated with a lower risk of all-cause mortality and cardiovascular mortality. There is now the need for a large randomized trial that compares the effect of a low vs high dialysate temperature on major cardiovascular outcomes. OBJECTIVE The purpose of this study is to test the effect of outpatient hemodialysis centers randomized to (1) a personalized temperature-reduced dialysate protocol or (2) a standard-temperature dialysate protocol for 4 years on cardiovascular-related death and hospitalizations. DESIGN The design of the study is a pragmatic, registry-based, open-label, cluster randomized controlled trial. SETTING Hemodialysis centers in Ontario, Canada, were randomized on February 1, 2017, for a trial start date of April 3, 2017, and end date of March 31, 2021. PARTICIPANTS In total, 84 hemodialysis centers will care for approximately 15 500 patients and provide over 4 million dialysis sessions over a 4-year follow-up. INTERVENTION Hemodialysis centers were randomized (1:1) to provide (1) a personalized temperature-reduced dialysate protocol or (2) a standard-temperature dialysate protocol of 36.5°C. For the personalized protocol, nurses set the dialysate temperature between 0.5°C and 0.9°C below the patient's predialysis body temperature for each dialysis session, to a minimum dialysate temperature of 35.5°C. PRIMARY OUTCOME A composite of cardiovascular-related death or major cardiovascular-related hospitalization (a hospital admission with myocardial infarction, congestive heart failure, or ischemic stroke) captured in Ontario health care administrative databases. PLANNED PRIMARY ANALYSIS The primary analysis will follow an intent-to-treat approach. The hazard ratio of time-to-first event will be estimated from a Cox model. Within-center correlation will be considered using a robust sandwich estimator. Observation time will be censored on the trial end date or when patients die from a noncardiovascular event. TRIAL REGISTRATION www.clinicaltrials.gov; identifier: NCT02628366.
Collapse
Affiliation(s)
- Ahmed A. Al-Jaishi
- London Health Sciences Centre, ON, Canada
- ICES, ON, Canada
- McMaster University, Hamilton, ON, Canada
| | | | - Jessica M. Sontrop
- London Health Sciences Centre, ON, Canada
- Western University, London, ON, Canada
| | - Stephanie N. Dixon
- London Health Sciences Centre, ON, Canada
- ICES, ON, Canada
- Western University, London, ON, Canada
| | | | | | | | - David Berry
- Sault Area Hospital, Sault Ste. Marie, ON, Canada
| | - Peter G. Blake
- London Health Sciences Centre, ON, Canada
- Western University, London, ON, Canada
| | | | | | | | | | | | | | | | - Jeremy M. Grimshaw
- Ottawa Hospital Research Institute, ON, Canada
- University of Ottawa, ON, Canada
| | | | | | - Arsh K. Jain
- London Health Sciences Centre, ON, Canada
- ICES, ON, Canada
- Western University, London, ON, Canada
| | | | - Reem A. Mustafa
- McMaster University, Hamilton, ON, Canada
- University of Kansas Medical Center, Kansas City, USA
| | | | | | - Matthew J. Oliver
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- University of Toronto, ON, Canada
| | | | | | | | - Justin Presseau
- Ottawa Hospital Research Institute, ON, Canada
- University of Ottawa, ON, Canada
| | - Eli Rabin
- Niagara Health System, St. Catharines, ON, Canada
| | | | | | - Manish M. Sood
- ICES, ON, Canada
- Ottawa Hospital Research Institute, ON, Canada
- University of Ottawa, ON, Canada
| | | | - Paul Tam
- Scarborough Health Network, ON, Canada
| | | | | | - Ron Wald
- ICES, ON, Canada
- University of Toronto, ON, Canada
- St. Michael’s Hospital, Toronto, ON, Canada
| | - Michael Walsh
- McMaster University, Hamilton, ON, Canada
- St. Joseph’s Healthcare, Hamilton, ON, Canada
| | - Paul Watson
- Thunder Bay Regional Health Sciences Centre, ON, Canada
| | | | | | | | - Amit X. Garg
- London Health Sciences Centre, ON, Canada
- ICES, ON, Canada
- McMaster University, Hamilton, ON, Canada
- Western University, London, ON, Canada
| |
Collapse
|
22
|
Chang LC, Sun HL, Tsai CH, Kuo CW, Liu KL, Lii CK, Huang CS, Li CC. 1,25(OH) 2 D 3 attenuates indoxyl sulfate-induced epithelial-to-mesenchymal cell transition via inactivation of PI3K/Akt/β-catenin signaling in renal tubular epithelial cells. Nutrition 2019; 69:110554. [PMID: 31536856 DOI: 10.1016/j.nut.2019.110554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/30/2019] [Accepted: 07/11/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Indoxyl sulfate (IS), a uremic toxin, has been shown to promote the epithelial-to-mesenchymal transition (EMT) of human proximal tubular cells and to accelerate the progression of chronic kidney disease (CKD). Despite the well-known protective role of 1,25-dihydroxyvitamin D3 [1,25(OH)2 D3] in EMT, the effect of 1,25(OH)2 D3 on IS-induced EMT in human proximal tubular epithelial cells and the underlying mechanism remain unclear. The aim of this study was to determine whether IS (0-1 mM) dose-dependently inhibited the protein expression of E-cadherin and increased the protein expression of alpha-smooth muscle actin, N-cadherin, and fibronectin. METHODS This study investigated the molecular mechanism by which 1,25(OH)2 D3 attenuates IS-induced EMT. HK-2 human renal tubular epithelial cells was used as the study model, and the MTT assay, Western Blotting, siRNA knockdown technique were used to explore the effects of 1,25(OH)2 D3 on EMT in the presence of IS. RESULTS Pretreatment with 1,25(OH)2 D3 inhibited the IS-induced EMT-associated protein expression in HK-2 cells. IS induced the phosphorylation of Akt (S473) and β-catenin (S552) and subsequently increased the nuclear accumulation of β-catenin. Pretreatment with 1,25(OH)2 D3 and LY294002 (phosphoinositide 3-kinase [PIK3] inhibitor) significantly inhibited the IS-induced phosphorylation of Akt and β-catenin, nuclear β-catenin accumulation, and EMT-associated protein expression. CONCLUSIONS Results from the present study revealed that the anti-EMT effect of 1,25(OH)2 D3 is likely through inhibition of the PI3K/Akt/β-catenin pathway, which leads to down-regulation of IS-driven EMT-associated protein expression in HK-2 human renal tubular epithelial cells.
Collapse
Affiliation(s)
- Li-Chien Chang
- Department of Internal Medicine, Taichung Armed Forces General Hospital, Taichung, Taiwan; Department of Medicine, National Defense Medical Center, Taipei, Taiwan; Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Hai-Lun Sun
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Han Tsai
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Wen Kuo
- Department of Internal Medicine, Taichung Armed Forces General Hospital, Taichung, Taiwan; Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Chin-Shiu Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
23
|
Remote sensing and signaling in kidney proximal tubules stimulates gut microbiome-derived organic anion secretion. Proc Natl Acad Sci U S A 2019; 116:16105-16110. [PMID: 31341083 PMCID: PMC6689987 DOI: 10.1073/pnas.1821809116] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Membrane transporters and receptors are responsible for balancing nutrient and metabolite levels to aid body homeostasis. Here, we report that proximal tubule cells in kidneys sense elevated endogenous, gut microbiome-derived, metabolite levels through EGF receptors and downstream signaling to induce their secretion by up-regulating the organic anion transporter-1 (OAT1). Remote metabolite sensing and signaling was observed in kidneys from healthy volunteers and rats in vivo, leading to induced OAT1 expression and increased removal of indoxyl sulfate, a prototypical microbiome-derived metabolite and uremic toxin. Using 2D and 3D human proximal tubule cell models, we show that indoxyl sulfate induces OAT1 via AhR and EGFR signaling, controlled by miR-223. Concomitantly produced reactive oxygen species (ROS) control OAT1 activity and are balanced by the glutathione pathway, as confirmed by cellular metabolomic profiling. Collectively, we demonstrate remote metabolite sensing and signaling as an effective OAT1 regulation mechanism to maintain plasma metabolite levels by controlling their secretion.
Collapse
|
24
|
Indoxyl Sulfate Induces Renal Fibroblast Activation through a Targetable Heat Shock Protein 90-Dependent Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2050183. [PMID: 31178953 PMCID: PMC6501427 DOI: 10.1155/2019/2050183] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/12/2019] [Indexed: 01/01/2023]
Abstract
Indoxyl sulfate (IS) accumulation occurs early during chronic kidney disease (CKD) progression and contributes to renal dysfunction by inducing fibrosis, inflammation, oxidative stress, and tissue remodeling. Renal toxicity of high IS concentrations (250 μM) has been widely explored, particularly in resident tubular and glomerular cells, while the effect of a moderate IS increase on kidneys is still mostly unknown. To define the effects of IS accumulation on renal fibroblasts, we first analyzed kidneys of C57BL/6 mice receiving IS (0.1%) in drinking water for 12 weeks. As a next step, we treated renal fibroblasts (NRK-49F) with IS (20 μM) with or without the HSP90 inhibitor 17-AAG (1 μM). In mouse kidneys, IS increased the collagen deposition and HSP90 and α-SMA expression (immunohistochemistry) in interstitial fibroblasts and caused tubular necrosis (histological H&E and picrosirius red staining). In NRK-49F cells, IS induced MCP1, TGF-β, collagen I, α-SMA, and HSP90 gene/protein expression and Smad2/3 pathway activation. IS had no effects on fibroblast proliferation and ROS production. 17-AAG counteracted IS-induced MCP1, TGF-β, collagen I, and α-SMA expression and Smad2/3 phosphorylation. Our study demonstrates that the IS increase promotes renal fibroblast activation by a HSP90-dependent pathway and indicates HSP90 inhibition as a potential strategy to restrain IS-induced kidney inflammation and fibrosis in CKD.
Collapse
|
25
|
Wang W, Hao G, Pan Y, Ma S, Yang T, Shi P, Zhu Q, Xie Y, Ma S, Zhang Q, Ruan H, Ding F. Serum indoxyl sulfate is associated with mortality in hospital-acquired acute kidney injury: a prospective cohort study. BMC Nephrol 2019; 20:57. [PMID: 30764800 PMCID: PMC6376694 DOI: 10.1186/s12882-019-1238-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 01/29/2019] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Protein-bound uremic toxins are associated with poor outcomes in patients with chronic kidney disease. The aim of this study is to investigate the relationship between indoxyl sulfate (IS), a protein-bound solute, and 90-day mortality in patients with acute kidney injury. METHODS Adults with hospital-acquired AKI (HA-AKI) were enrolled in this prospective cohort study between 2014 and 2015, according to the KDIGO creatinine criteria. The primary end point was all-cause death during follow-up. RESULTS The mean serum IS level in patients with HA-AKI was 2.74 ± 0.75 μg/ml, which was higher than that in healthy subjects (1.73 ± 0.11 μg/ml, P < 0.001) and critically ill patients (2.46 ± 0.35 μg/ml, P = 0.016) but was lower than that in patients with chronic kidney disease (3.07 ± 0.31 μg/ml, P < 0.001). Furthermore, serum IS levels (2.83 ± 0.55 μg/ml) remained elevated in patients with HA-AKI on the seventh day after AKI diagnosis. Patients with HA-AKI were divided into the following two groups according to the median serum IS level: the low-IS group and the high-IS group. A total of 94 (35.9%) patient deaths occurred within 90 days, including 76 (29.0%) in the low-IS group and 112 (42.7%) in the high-IS group (P = 0.019). Kaplan-Meier analysis revealed that the two groups differed significantly with respect to 90-day survival (log-rank P = 0.007), and Cox regression analysis showed that an IS level ≥ 2.74 μg/ml was significantly associated with a 2.0-fold increased risk of death (adjusted hazard ratio [HR], 2.92; 95% confidence interval [CI], 1.76 to 4.86; P < 0.001) compared with an IS level < 2.74 μg/ml. CONCLUSIONS Serum IS levels were significantly elevated in patients with HA-AKI compared to those in healthy subjects and critically ill patients and were associated with a worse prognosis of HA-AKI. TRIAL REGISTRATION www.clinicaltrials.gov NCT 00953992. Registered 6 August 2009.
Collapse
Affiliation(s)
- Wenji Wang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Guihua Hao
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yu Pan
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Shuai Ma
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Tianye Yang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Peng Shi
- Department of Medical Statistics, Children's Hospital; Center for Evidence-based Medicine, Fudan University, Shanghai, 200433, China
| | - Qiuyu Zhu
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yingxin Xie
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Shaojun Ma
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Qi Zhang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Hong Ruan
- Department of Nursing, Clinical Medical School, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Feng Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
26
|
Contributory Role of Gut Microbiota and Their Metabolites Toward Cardiovascular Complications in Chronic Kidney Disease. Semin Nephrol 2019; 38:193-205. [PMID: 29602401 DOI: 10.1016/j.semnephrol.2018.01.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The gut microbiome recently has emerged as a novel risk factor that impacts health and disease. Our gut microbiota can function as an endocrine organ through its unique ability to metabolize various dietary precursors, and can fuel the systemic inflammation observed in chronic disease. This is especially important in the setting of chronic kidney disease, in which microbial metabolism can contribute directly to accumulation of circulating toxins that then can alter and shift the balance of microbiota composition and downstream functions. To study this process, advances in -omics technologies are providing opportunities to understand not only the taxonomy, but also the functional diversity of our microbiome. We also reliably can quantify en masse a wide range of uremic byproducts of microbial metabolism. Herein, we examine the bidirectional relationship between the gut microbiome and the failing kidneys. We describe potential approaches targeting gut microbiota for cardiovascular risk reduction in chronic kidney disease using an illustrative example of a novel gut-generated metabolite, trimethylamine N-oxide.
Collapse
|
27
|
Role of Uremic Toxins for Kidney, Cardiovascular, and Bone Dysfunction. Toxins (Basel) 2018; 10:toxins10050202. [PMID: 29772660 PMCID: PMC5983258 DOI: 10.3390/toxins10050202] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/04/2018] [Accepted: 05/10/2018] [Indexed: 02/07/2023] Open
Abstract
With decreasing kidney function, cardiovascular disease (CVD) and mineral bone disorders frequently emerge in patients with chronic kidney disease (CKD). For these patients, in addition to the traditional risk factors, non-traditional CKD-specific risk factors are also associated with such diseases and conditions. One of these non-traditional risk factors is the accumulation of uremic toxins (UTs). In addition, the accumulation of UTs further deteriorates kidney function. Recently, a huge number of UTs have been identified. Although many experimental and clinical studies have reported associations between UTs and the progression of CKD, CVD, and bone disease, these relationships are very complex and have not been fully elucidated. Among the UTs, indoxyl sulfate, asymmetric dimethylarginine, and p-cresylsulfate have been of particular focus, up until now. In this review, we summarize the pathophysiological influences of these UTs on the kidney, cardiovascular system, and bone, and discuss the clinical data regarding the harmful effects of these UTs on diseases and conditions.
Collapse
|
28
|
Naseeb U, Zarina S, Jägerbrink T, Shafqat J, Jörnvall H, Axelsson J. Differential hemoglobin A sequestration between hemodialysis modalities. Biomol Concepts 2018; 8:125-129. [PMID: 28422703 DOI: 10.1515/bmc-2017-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/09/2017] [Indexed: 11/15/2022] Open
Abstract
This report evaluates plasma protein patterns, dialysates and protein analysis of used dialysis membranes from the same patient under hemodialysis in three separate modalities, using high-flux membranes in concentration-driven transport (HD), convection-driven hemofiltration (HF) and combined hemodialfiltration (HDF). The plasma protein changes induced by each of the three dialysis modalities showed small differences in proteins identified towards our previous plasma analyses of chronic kidney disease (CKD) patients. The used dialysate peptide concentrations likewise exhibited small differences among the modalities and varied in the same relative order as the plasma changes, with protein losses in the order HD>HDF>HF. The membrane protein deposits allowed quantification of the relative Hb removal ratios as ~1.7 for HD and ~1.2 for HDF vs. ~1.0 for HF. Hence, plasma protein alterations, dialysate peptide contents and membrane Hb deposits all identify HD as the modality with the most extensive filtration results and exemplifies the accessibility of protein analysis of used membrane filters for evaluation of dialysis efficiencies.
Collapse
|
29
|
Enoki Y, Watanabe H, Arake R, Sugimoto R, Imafuku T, Tominaga Y, Ishima Y, Kotani S, Nakajima M, Tanaka M, Matsushita K, Fukagawa M, Otagiri M, Maruyama T. Indoxyl sulfate potentiates skeletal muscle atrophy by inducing the oxidative stress-mediated expression of myostatin and atrogin-1. Sci Rep 2016; 6:32084. [PMID: 27549031 PMCID: PMC4994088 DOI: 10.1038/srep32084] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 08/02/2016] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle atrophy, referred to as sarcopenia, is often observed in chronic kidney disease (CKD) patients, especially in patients who are undergoing hemodialysis. The purpose of this study was to determine whether uremic toxins are involved in CKD-related skeletal muscle atrophy. Among six protein-bound uremic toxins, indole containing compounds, indoxyl sulfate (IS) significantly inhibited proliferation and myotube formation in C2C12 myoblast cells. IS increased the factors related to skeletal muscle breakdown, such as reactive oxygen species (ROS) and inflammatory cytokines (TNF-α, IL-6 and TGF-β1) in C2C12 cells. IS also enhanced the production of muscle atrophy-related genes, myostatin and atrogin-1. These effects induced by IS were suppressed in the presence of an antioxidant or inhibitors of the organic anion transporter and aryl hydrocarbon receptor. The administered IS was distributed to skeletal muscle and induced superoxide production in half-nephrectomized (1/2 Nx) mice. The chronic administration of IS significantly reduced the body weights accompanied by skeletal muscle weight loss. Similar to the in vitro data, IS induced the expression of myostatin and atrogin-1 in addition to increasing the production of inflammatory cytokines by enhancing oxidative stress in skeletal muscle. These data suggest that IS has the potential to accelerate skeletal muscle atrophy by inducing oxidative stress-mediated myostatin and atrogin-1 expression.
Collapse
Affiliation(s)
- Yuki Enoki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan.,Center for Clinical Pharmaceutical Sciences, School of Pharmacy, Kumamoto University, Kumamoto, Japan
| | - Riho Arake
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan
| | - Ryusei Sugimoto
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan
| | - Tadashi Imafuku
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan
| | - Yuna Tominaga
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan
| | - Yu Ishima
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan.,Center for Clinical Pharmaceutical Sciences, School of Pharmacy, Kumamoto University, Kumamoto, Japan
| | - Shunsuke Kotani
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Makoto Nakajima
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Motoko Tanaka
- Department of Nephrology, Akebono Clinic, Kumamoto, Japan
| | | | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Kanagawa, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Kumamoto, Japan.,DDS Research Institute, Sojo University, Kumamoto, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Japan.,Center for Clinical Pharmaceutical Sciences, School of Pharmacy, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
30
|
Zhang LS, Davies SS. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med 2016; 8:46. [PMID: 27102537 PMCID: PMC4840492 DOI: 10.1186/s13073-016-0296-x] [Citation(s) in RCA: 348] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mass spectrometry- and nuclear magnetic resonance-based metabolomic studies comparing diseased versus healthy individuals have shown that microbial metabolites are often the compounds most markedly altered in the disease state. Recent studies suggest that several of these metabolites that derive from microbial transformation of dietary components have significant effects on physiological processes such as gut and immune homeostasis, energy metabolism, vascular function, and neurological behavior. Here, we review several of the most intriguing diet-dependent metabolites that may impact host physiology and may therefore be appropriate targets for therapeutic interventions, such as short-chain fatty acids, trimethylamine N-oxide, tryptophan and tyrosine derivatives, and oxidized fatty acids. Such interventions will require modulating either bacterial species or the bacterial biosynthetic enzymes required to produce these metabolites, so we briefly describe the current understanding of the bacterial and enzymatic pathways involved in their biosynthesis and summarize their molecular mechanisms of action. We then discuss in more detail the impact of these metabolites on health and disease, and review current strategies to modulate levels of these metabolites to promote human health. We also suggest future studies that are needed to realize the full therapeutic potential of targeting the gut microbiota.
Collapse
Affiliation(s)
- Linda S Zhang
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Sean S Davies
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA. .,Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA. .,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
31
|
Gajjala PR, Sanati M, Jankowski J. Cellular and Molecular Mechanisms of Chronic Kidney Disease with Diabetes Mellitus and Cardiovascular Diseases as Its Comorbidities. Front Immunol 2015. [PMID: 26217336 PMCID: PMC4495338 DOI: 10.3389/fimmu.2015.00340] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD), diabetes mellitus (DM), and cardiovascular diseases (CVD) are complex disorders of partly unknown genesis and mostly known progression factors. CVD and DM are the risk factors of CKD and are strongly intertwined since DM can lead to both CKD and/or CVD, and CVD can lead to kidney disease. In recent years, our knowledge of CKD, DM, and CVD has been expanded and several important experimental, clinical, and epidemiological associations have been reported. The tight cellular and molecular interactions between the renal, diabetic, and cardiovascular systems in acute or chronic disease settings are becoming increasingly evident. However, the (patho-) physiological basis of the interactions of CKD, DM, and CVD with involvement of multiple endogenous and environmental factors is highly complex and our knowledge is still at its infancy. Not only single pathways and mediators of progression of these diseases have to be considered in these processes but also the mutual interactions of these factors are essential. The recent advances in proteomics and integrative analysis technologies have allowed rapid progress in analyzing complex disorders and clearly show the opportunity for new efficient and specific therapies. More than a dozen pathways have been identified so far, including hyperactivity of the renin–angiotensin (RAS)–aldosterone system, osmotic sodium retention, endothelial dysfunction, dyslipidemia, RAS/RAF/extracellular-signal-regulated kinase pathway, modification of the purinergic system, phosphatidylinositol 3-kinase (PI 3-kinase)-dependent signaling pathways, and inflammation, all leading to histomorphological alterations of the kidney and vessels of diabetic and non-diabetic patients. Since a better understanding of the common cellular and molecular mechanisms of these diseases may be a key to successful identification of new therapeutic targets, we review in this paper the current literature about cellular and molecular mechanisms of CKD.
Collapse
Affiliation(s)
- Prathibha Reddy Gajjala
- Institute for Molecular Cardiovascular Research, Universitätsklinikum RWTH Aachen , Aachen , Germany
| | - Maryam Sanati
- Institute for Molecular Cardiovascular Research, Universitätsklinikum RWTH Aachen , Aachen , Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, Universitätsklinikum RWTH Aachen , Aachen , Germany
| |
Collapse
|
32
|
Glorieux G, Tattersall J. Uraemic toxins and new methods to control their accumulation: game changers for the concept of dialysis adequacy. Clin Kidney J 2015; 8:353-62. [PMID: 26251699 PMCID: PMC4515890 DOI: 10.1093/ckj/sfv034] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 02/07/2023] Open
Abstract
The current concept of an adequate dialysis based only on the dialysis process itself is rather limited. We now have considerable knowledge of uraemic toxicity and improved tools for limiting uraemic toxin accumulation. It is time to make use of these. A broader concept of adequacy that focusses on uraemic toxicity is required. As discussed in the present review, adequacy could be achieved by many different methods in combination with, or instead of, dialysis. These include preservation of renal function, dietary intake, reducing uraemic toxin generation rate and intestinal absorption, isolated ultrafiltration and extracorporeal adsorption of key uraemic toxins. A better measure of the quality of dialysis treatment would quantify the uraemic state in the patient using levels of a panel of key uraemic toxins. Treatment would focus on controlling uraemic toxicity while reducing harm or inconvenience to the patient. Delivering more dialysis might not be the best way to achieve this.
Collapse
Affiliation(s)
- Griet Glorieux
- Department of Internal Medicine, Nephrology Division , Ghent University Hospital , Gent , Belgium
| | - James Tattersall
- Department of Renal Medicine , Leeds Teaching Hospitals , Leeds LS2 7EF , UK
| |
Collapse
|
33
|
Young GH, Lin JT, Cheng YF, Huang CF, Chao CY, Nong JY, Chen PK, Chen HM. Identification of adenine modulating AMPK activation in NIH/3T3 cells by proteomic approach. J Proteomics 2015; 120:204-14. [DOI: 10.1016/j.jprot.2015.03.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 02/28/2015] [Accepted: 03/12/2015] [Indexed: 02/08/2023]
|
34
|
Vanholder R, Glorieux G. The intestine and the kidneys: a bad marriage can be hazardous. Clin Kidney J 2015; 8:168-79. [PMID: 25815173 PMCID: PMC4370304 DOI: 10.1093/ckj/sfv004] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
The concept that the intestine and chronic kidney disease influence each other, emerged only recently. The problem is multifaceted and bidirectional. On one hand, the composition of the intestinal microbiota impacts uraemic retention solute production, resulting in the generation of essentially protein-bound uraemic toxins with strong biological impact such as vascular damage and progression of kidney failure. On the other hand, the uraemic status affects the composition of intestinal microbiota, the generation of uraemic retention solutes and their precursors and causes disturbances in the protective epithelial barrier of the intestine and the translocation of intestinal microbiota into the body. All these elements together contribute to the disruption of the metabolic equilibrium and homeostasis typical to uraemia. Several measures with putative impact on intestinal status have recently been tested for their influence on the generation or concentration of uraemic toxins. These include dietary measures, prebiotics, probiotics, synbiotics and intestinal sorbents. Unfortunately, the quality and the evidence base of many of these studies are debatable, especially in uraemia, and often results within one study or among studies are contradictory. Nevertheless, intestinal uraemic metabolite generation remains an interesting target to obtain in the future as an alternative or additive to dialysis to decrease uraemic toxin generation. In the present review, we aim to summarize (i) the role of the intestine in uraemia by producing uraemic toxins and by generating pathophysiologically relevant changes, (ii) the role of uraemia in modifying intestinal physiology and (iii) the therapeutic options that could help to modify these effects and the studies that have assessed the impact of these therapies.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, 0K12 , University Hospital , Ghent B9000 , Belgium
| | - Griet Glorieux
- Nephrology Section, 0K12 , University Hospital , Ghent B9000 , Belgium
| |
Collapse
|