1
|
Zakrocka I, Urbańska EM, Załuska W, Kronbichler A. Kynurenine Pathway after Kidney Transplantation: Friend or Foe? Int J Mol Sci 2024; 25:9940. [PMID: 39337426 PMCID: PMC11432217 DOI: 10.3390/ijms25189940] [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: 08/21/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Kidney transplantation significantly improves the survival of patients with end-stage kidney disease (ESKD) compared to other forms of kidney replacement therapy. However, kidney transplant recipients' outcomes are not fully satisfactory due to increased risk of cardiovascular diseases, infections, and malignancies. Immune-related complications remain the biggest challenge in the management of kidney graft recipients. Despite the broad spectrum of immunosuppressive agents available and more detailed methods used to monitor their effectiveness, chronic allograft nephropathy remains the most common cause of kidney graft rejection. The kynurenine (KYN) pathway is the main route of tryptophan (Trp) degradation, resulting in the production of a plethora of substances with ambiguous properties. Conversion of Trp to KYN by the enzyme indoleamine 2,3-dioxygenase (IDO) is the rate-limiting step determining the formation of the next agents from the KYN pathway. IDO activity, as well as the production of subsequent metabolites of the pathway, is highly dependent on the balance between pro- and anti-inflammatory conditions. Moreover, KYN pathway products themselves possess immunomodulating properties, e.g., modify the activity of IDO and control other immune-related processes. KYN metabolites were widely studied in neurological disorders but recently gained the attention of researchers in the context of immune-mediated diseases. Evidence that this route of Trp degradation may represent a peripheral tolerogenic pathway with significant implications for transplantation further fueled this interest. Our review aimed to present recent knowledge about the role of the KYN pathway in the pathogenesis, diagnosis, monitoring, and treatment of kidney transplant recipients' complications.
Collapse
Affiliation(s)
- Izabela Zakrocka
- Department of Nephrology, Medical University of Lublin, 20-093 Lublin, Poland; (I.Z.); (W.Z.)
| | - Ewa M. Urbańska
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Wojciech Załuska
- Department of Nephrology, Medical University of Lublin, 20-093 Lublin, Poland; (I.Z.); (W.Z.)
| | - Andreas Kronbichler
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, 6020 Innsbruck, Austria
- Department of Health, Medicine and Caring Sciences, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
2
|
Nagy-Grócz G, Spekker E, Vécsei L. Kynurenines, Neuronal Excitotoxicity, and Mitochondrial Oxidative Stress: Role of the Intestinal Flora. Int J Mol Sci 2024; 25:1698. [PMID: 38338981 PMCID: PMC10855176 DOI: 10.3390/ijms25031698] [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: 01/04/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
The intestinal flora has been the focus of numerous investigations recently, with inquiries not just into the gastrointestinal aspects but also the pathomechanism of other diseases such as nervous system disorders and mitochondrial diseases. Mitochondrial disorders are the most common type of inheritable metabolic illness caused by mutations of mitochondrial and nuclear DNA. Despite the intensive research, its diagnosis is usually difficult, and unfortunately, treating it challenges physicians. Metabolites of the kynurenine pathway are linked to many disorders, such as depression, schizophrenia, migraine, and also diseases associated with impaired mitochondrial function. The kynurenine pathway includes many substances, for instance kynurenic acid and quinolinic acid. In this review, we would like to show a possible link between the metabolites of the kynurenine pathway and mitochondrial stress in the context of intestinal flora. Furthermore, we summarize the possible markers of and future therapeutic options for the kynurenine pathway in excitotoxicity and mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Gábor Nagy-Grócz
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary;
- Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| | | | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary;
- HUN-REN-SZTE Neuroscience Research Group, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
3
|
El Chamieh C, Larabi IA, Alencar De Pinho N, Lambert O, Combe C, Fouque D, Frimat L, Jacquelinet C, Laville M, Laville S, Lange C, Alvarez JC, Massy ZA, Liabeuf S. Study of the association between serum levels of kynurenine and cardiovascular outcomes and overall mortality in chronic kidney disease. Clin Kidney J 2024; 17:sfad248. [PMID: 38186868 PMCID: PMC10768787 DOI: 10.1093/ckj/sfad248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Indexed: 01/09/2024] Open
Abstract
Background Kynurenine is a protein-bound uremic toxin. Its circulating levels are increased in chronic kidney disease (CKD). Experimental studies showed that it exerted deleterious cardiovascular effects. We sought to evaluate an association between serum kynurenine levels and adverse fatal or nonfatal cardiovascular events and all-cause mortality in CKD patients. Methods The CKD-REIN study is a prospective cohort of people with CKD having an estimated glomerular filtration rate (eGFR) <60 ml/min/1.73 m². Baseline frozen samples of total and free fractions of kynurenine and tryptophan were measured using a validated liquid chromatography tandem mass spectrometry technique. Cause-specific Cox models were used to estimate hazard ratios (HRs) for each outcome. Results Of the 2406 included patients (median age: 68 years; median eGFR: 25 ml/min/1.73 m2), 52% had a history of cardiovascular disease. A doubling of serum-free kynurenine levels was associated with an 18% increased hazard of cardiovascular events [466 events, HR (95%CI):1.18(1.02,1.33)], independently of eGFR, serum-free tryptophan level or other uremic toxins, cardioprotective drugs, and traditional cardiovascular risk factors. Serum-free kynurenine was significantly associated with non-atheromatous cardiovascular events [HR(95%CI):1.26(1.03,1.50)], but not with atheromatous cardiovascular events [HR(95%CI):1.15(0.89,1.50)]. The association of serum-free kynurenine with cardiovascular mortality was also independently significant [87 events; adjusted HR(95%CI):1.64(1.10,2.40)]. However, the association of serum-free kynurenine with all-cause mortality was no more significant after adjustment on serum-free tryptophan [311 events, HR(95%CI):1.12(0.90, 1.40)]. Conclusions Our findings imply that serum-free kynurenine, independently of other cardiovascular risk factors (including eGFR), is associated with fatal or nonfatal cardiovascular outcomes, particularly non-atheromatous cardiovascular events; in patients with CKD. Strategies to reduce serum kynurenine levels should be evaluated in further studies.
Collapse
Affiliation(s)
- Carolla El Chamieh
- Centre for Research in Epidemiology and Population Health (CESP), INSERM UMRS 1018, Université Paris-Saclay, Université Versailles Saint Quentin, Villejuif, France
| | - Islam Amine Larabi
- Department of Pharmacology and Toxicology, Raymond Poincaré Hospital, AP-HP, Garches, France
- UVSQ, Université Paris-Saclay, Inserm U1018, CESP, Équipe MOODS, MasSpecLab, Montigny-le-Bretonneux, France
| | - Natalia Alencar De Pinho
- Centre for Research in Epidemiology and Population Health (CESP), INSERM UMRS 1018, Université Paris-Saclay, Université Versailles Saint Quentin, Villejuif, France
| | - Oriane Lambert
- Centre for Research in Epidemiology and Population Health (CESP), INSERM UMRS 1018, Université Paris-Saclay, Université Versailles Saint Quentin, Villejuif, France
| | - Christian Combe
- Service de Néphrologie Transplantation Dialyse Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
- INSERM, U1026, University Bordeaux, Bordeaux, France
| | - Denis Fouque
- Nephrology Dept, Centre Hospitalier Lyon Sud, Université de Lyon, Carmen, Pierre-Bénite, France
- Université de Lyon, CarMeN INSERM 1060, Lyon, France
| | - Luc Frimat
- Nephrology Department, CHRU de Nancy, Vandoeuvre-lès-Nancy, France
- Lorraine University, APEMAC, Vandoeuvre-lès-Nancy, France
| | - Christian Jacquelinet
- Centre for Research in Epidemiology and Population Health (CESP), INSERM UMRS 1018, Université Paris-Saclay, Université Versailles Saint Quentin, Villejuif, France
- Biomedecine Agency, Saint Denis La Plaine, France
| | | | - Solène Laville
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens-Picardie University Medical Center, Amiens, France
- MP3CV Laboratory, Jules Verne University of Picardie, Amiens, France
| | - Céline Lange
- Centre for Research in Epidemiology and Population Health (CESP), INSERM UMRS 1018, Université Paris-Saclay, Université Versailles Saint Quentin, Villejuif, France
| | - Jean-Claude Alvarez
- Department of Pharmacology and Toxicology, Raymond Poincaré Hospital, AP-HP, Garches, France
- UVSQ, Université Paris-Saclay, Inserm U1018, CESP, Équipe MOODS, MasSpecLab, Montigny-le-Bretonneux, France
| | - Ziad A Massy
- Centre for Research in Epidemiology and Population Health (CESP), INSERM UMRS 1018, Université Paris-Saclay, Université Versailles Saint Quentin, Villejuif, France
- Department of Nephrology, Ambroise Paré University Hospital, APHP, Boulogne-Billancourt, France
| | - Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens-Picardie University Medical Center, Amiens, France
- MP3CV Laboratory, Jules Verne University of Picardie, Amiens, France
| |
Collapse
|
4
|
Mörtberg J, Salzinger B, Lundwall K, Edfors R, Jacobson SH, Wallén HN, Jernberg T, Baron T, Erlinge D, Andell P, James S, Eggers KM, Hjort M, Kahan T, Lundman P, Tornvall P, Rezeli M, Marko-Varga G, Lindahl B, Spaak J. Prognostic importance of biomarkers associated with haemostatic, vascular and endothelial disturbances in acute coronary syndrome patients in relation to kidney function. Int J Cardiol 2023; 373:64-71. [PMID: 36476672 DOI: 10.1016/j.ijcard.2022.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/15/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Patients with kidney failure have a high risk for cardiovascular events. We aimed to evaluate the prognostic importance of selected biomarkers related to haemostasis, endothelial function, and vascular regulation in patients with acute coronary syndrome (ACS), and to study whether this association differed in patients with renal dysfunction. METHODS Plasma was collected in 1370 ACS patients included between 2008 and 2015. Biomarkers were analysed using a Proximity Extension Assay and a Multiple Reaction Monitoring mass spectrometry assay. To reduce multiplicity, biomarkers correlating with eGFR were selected a priori among 36 plasma biomarkers reflecting endothelial and vascular function, and haemostasis. Adjusted Cox regression were used to study their association with the composite outcome of myocardial infarction, ischemic stroke, heart failure or death. Interaction with eGFR strata above or below 60 ml/min/1.73 m2 was tested. RESULTS Tissue factor, proteinase-activated receptor, soluble urokinase plasminogen activator surface receptor (suPAR), thrombomodulin, adrenomedullin, renin, and angiotensinogen correlated inversely with eGFR and were selected for the Cox regression. Mean follow-up was 5.2 years during which 428 events occurred. Adrenomedullin, suPAR, and renin were independently associated with the composite outcome. Adrenomedullin showed interaction with eGFR strata (p = 0.010) and was associated with increased risk (HR 1.88; CI 1.44-2.45) only in patients with eGFR ≥60 ml/min/ 1.73 m2. CONCLUSIONS Adrenomedullin, suPAR, and renin were associated with the composite outcome in all. Adrenomedullin, involved in endothelial protection, showed a significant interaction with renal function and outcome, and was associated with the composite outcome only in patients with preserved kidney function.
Collapse
Affiliation(s)
- Josefin Mörtberg
- Centre for Clinical Research, Vastmanland Hospital Vasteras, Region Vastmanland - Uppsala University, Sweden; Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Barbara Salzinger
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Lundwall
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Robert Edfors
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Stefan H Jacobson
- Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Håkan N Wallén
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Jernberg
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Tomasz Baron
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - David Erlinge
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Pontus Andell
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden
| | - Stefan James
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Kai M Eggers
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Marcus Hjort
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Thomas Kahan
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Pia Lundman
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Per Tornvall
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Melinda Rezeli
- Clinical Protein Science & Imaging, Department of Biomedical Engineering, Lund University, BMC D13, Lund SE-221 84, Sweden
| | - György Marko-Varga
- Clinical Protein Science & Imaging, Department of Biomedical Engineering, Lund University, BMC D13, Lund SE-221 84, Sweden
| | - Bertil Lindahl
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Jonas Spaak
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Oe Y, Takahashi N. Tissue Factor, Thrombosis, and Chronic Kidney Disease. Biomedicines 2022; 10:2737. [PMID: 36359257 PMCID: PMC9687479 DOI: 10.3390/biomedicines10112737] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/14/2023] Open
Abstract
Coagulation abnormalities are common in chronic kidney disease (CKD). Tissue factor (TF, factor III) is a master regulator of the extrinsic coagulation system, activating downstream coagulation proteases, such as factor Xa and thrombin, and promoting fibrin formation. TF and coagulation proteases also activate protease-activated receptors (PARs) and are implicated in various organ injuries. Recent studies have shown the mechanisms by which thrombotic tendency is increased under CKD-specific conditions. Uremic toxins, such as indoxyl sulfate and kynurenine, are accumulated in CKD and activate TF and coagulation; in addition, the TF-coagulation protease-PAR pathway enhances inflammation and fibrosis, thereby exacerbating renal injury. Herein, we review the recent research studies to understand the role of TF in increasing the thrombotic risk and CKD progression.
Collapse
Affiliation(s)
- Yuji Oe
- Division of Nephrology, Rheumatology, and Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA 92161, USA
- VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Nobuyuki Takahashi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Sendai 980-8578, Japan
| |
Collapse
|
6
|
Ala M, Eftekhar SP. The Footprint of Kynurenine Pathway in Cardiovascular Diseases. Int J Tryptophan Res 2022; 15:11786469221096643. [PMID: 35784899 PMCID: PMC9248048 DOI: 10.1177/11786469221096643] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/28/2022] [Indexed: 12/30/2022] Open
Abstract
Kynurenine pathway is the main route of tryptophan metabolism and produces several metabolites with various biologic properties. It has been uncovered that several cardiovascular diseases are associated with the overactivation of kynurenine pathway and kynurenine and its metabolites have diagnostic and prognostic value in cardiovascular diseases. Furthermore, it was found that several kynurenine metabolites can differently affect cardiovascular health. For instance, preclinical studies have shown that kynurenine, xanthurenic acid and cis-WOOH decrease blood pressure; kynurenine and 3-hydroxyanthranilic acid prevent atherosclerosis; kynurenic acid supplementation and kynurenine 3-monooxygenase (KMO) inhibition improve the outcome of stroke. Indoleamine 2,3-dioxygenase (IDO) overactivity and increased kynurenine levels improve cardiac and vascular transplantation outcomes, whereas exacerbating the outcome of myocardial ischemia, post-ischemic myocardial remodeling, and abdominal aorta aneurysm. IDO inhibition and KMO inhibition are also protective against viral myocarditis. In addition, dysregulation of kynurenine pathway is observed in several conditions such as senescence, depression, diabetes, chronic kidney disease (CKD), cirrhosis, and cancer closely connected to cardiovascular dysfunction. It is worth defining the exact effect of each metabolite of kynurenine pathway on cardiovascular health. This narrative review is the first review that separately discusses the involvement of kynurenine pathway in different cardiovascular diseases and dissects the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Seyed Parsa Eftekhar
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
7
|
Abstract
Kidney diseases have become one of the most common health care problems. Due to a growing number of advanced aged patients with concomitant disorders the prevalence of these diseases will increase over the coming decades. Despite available laboratory tests, accurate and rapid diagnosis of renal dysfunction has yet to be realized, and prognosis is uncertain. Moreover, data on diagnostic and prognostic markers in kidney diseases are lacking. The kynurenine (KYN) pathway is one of the routes of tryptophan (Trp) degradation, with biologically active substances presenting ambiguous properties. The KYN pathway is known to be highly dependent on immunological system activity. As the kidneys are one of the main organs involved in the formation, degradation and excretion of Trp end products, pathologies involving the kidneys result in KYN pathway activity disturbances. This review aims to summarize changes in the KYN pathway observed in the most common kidney disease, chronic kidney disease (CKD), with a special focus on diabetic kidney disease, acute kidney injury (AKI), glomerulonephritis and kidney graft function monitoring. Additionally, the importance of KYN pathway activity in kidney cancer pathogenesis is discussed, as are available pharmacological agents affecting KYN pathway activity in the kidney. Despite limited clinical data, the KYN pathway appears to be a promising target in the diagnosis and prognosis of kidney diseases. Modulation of KYN pathway activity by pharmacological agents should be considered in the treatment of kidney diseases.
Collapse
|
8
|
Wee HN, Liu JJ, Ching J, Kovalik JP, Lim SC. The Kynurenine Pathway in Acute Kidney Injury and Chronic Kidney Disease. Am J Nephrol 2021; 52:771-787. [PMID: 34753140 PMCID: PMC8743908 DOI: 10.1159/000519811] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The kynurenine pathway (KP) is the major catabolic pathway for tryptophan degradation. The KP plays an important role as the sole de novo nicotinamide adenine dinucleotide (NAD+) biosynthetic pathway in normal human physiology and functions as a counter-regulatory mechanism to mitigate immune responses during inflammation. Although the KP has been implicated in a variety of disorders including Huntington's disease, seizures, cardiovascular disease, and osteoporosis, its role in renal diseases is seldom discussed. SUMMARY This review summarizes the roles of the KP and its metabolites in acute kidney injury (AKI) and chronic kidney disease (CKD) based on current literature evidence. Metabolomics studies demonstrated that the KP metabolites were significantly altered in patients and animal models with AKI or CKD. The diagnostic and prognostic values of the KP metabolites in AKI and CKD were highlighted in cross-sectional and longitudinal human observational studies. The biological impact of the KP on the pathophysiology of AKI and CKD has been studied in experimental models of different etiologies. In particular, the activation of the KP was found to confer protection in animal models of glomerulonephritis, and its immunomodulatory mechanism may involve the regulation of T cell subsets such as Th17 and regulatory T cells. Manipulation of the KP to increase NAD+ production or diversion toward specific KP metabolites was also found to be beneficial in animal models of AKI. Key Messages: KP metabolites are reported to be dysregulated in human observational and animal experimental studies of AKI and CKD. In AKI, the magnitude and direction of changes in the KP depend on the etiology of the damage. In CKD, KP metabolites are altered with the onset and progression of CKD all the way to advanced stages of the disease, including uremia and its related vascular complications. The activation of the KP and diversion to specific sub-branches are currently being explored as therapeutic strategies in these diseases, especially with regards to the immunomodulatory effects of certain KP metabolites. Further elucidation of the KP may hold promise for the development of biomarkers and targeted therapies for these kidney diseases.
Collapse
Affiliation(s)
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Jianhong Ching
- Duke-NUS Medical School, Singapore, Singapore
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | | | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Diabetes Centre, Admiralty Medical Centre, Singapore, Singapore
| |
Collapse
|
9
|
Thrombolome and Its Emerging Role in Chronic Kidney Diseases. Toxins (Basel) 2021; 13:toxins13030223. [PMID: 33803899 PMCID: PMC8003125 DOI: 10.3390/toxins13030223] [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: 02/07/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at an increased risk of thromboembolic complications, including myocardial infarction, stroke, deep vein thrombosis, and pulmonary embolism. These complications lead to increased mortality. Evidence points to the key role of CKD-associated dysbiosis and its effect via the generation of gut microbial metabolites in inducing the prothrombotic phenotype. This phenomenon is known as thrombolome, a panel of intestinal bacteria-derived uremic toxins that enhance thrombosis via increased tissue factor expression, platelet hyperactivity, microparticles release, and endothelial dysfunction. This review discusses the role of uremic toxins derived from gut-microbiota metabolism of dietary tryptophan (indoxyl sulfate (IS), indole-3-acetic acid (IAA), kynurenine (KYN)), phenylalanine/tyrosine (p-cresol sulfate (PCS), p-cresol glucuronide (PCG), phenylacetylglutamine (PAGln)) and choline/phosphatidylcholine (trimethylamine N-oxide (TMAO)) in spontaneously induced thrombosis. The increase in the generation of gut microbial uremic toxins, the activation of aryl hydrocarbon (AhRs) and platelet adrenergic (ARs) receptors, and the nuclear factor kappa B (NF-κB) signaling pathway can serve as potential targets during the prevention of thromboembolic events. They can also help create a new therapeutic approach in the CKD population.
Collapse
|
10
|
Molecular Mechanisms Underlying the Cardiovascular Toxicity of Specific Uremic Solutes. Cells 2020; 9:cells9092024. [PMID: 32887404 PMCID: PMC7565564 DOI: 10.3390/cells9092024] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Mounting evidence strongly suggests a causal link between chronic kidney disease (CKD) and cardiovascular disease (CVD). Compared with non-CKD patients, patients with CKD suffer disproportionately from CVD and derive suboptimal benefits from interventions targeting conventional CVD risk factors. Uremic toxins (UTs), whose plasma levels rapidly rise as CKD progresses, represent a unique risk factor in CKD, which has protean manifestations on CVD. Among the known UTs, tryptophan metabolites and trimethylamine N-oxide are well-established cardiovascular toxins. Their molecular mechanisms of effect warrant special consideration to draw translational value. This review surveys current knowledge on the effects of specific UTs on different pathways and cell functions that influence the integrity of cardiovascular health, with implication for CVD progression. The effect of UTs on cardiovascular health is an example of a paradigm in which a cascade of molecular and metabolic events induced by pathology in one organ in turn induces dysfunction in another organ. Deciphering the molecular mechanisms underlying such cross-organ pathologies will help uncover therapeutic targets to improve the management of CVD in patients with CKD.
Collapse
|
11
|
Yu J, Lin T, Huang N, Xia X, Li J, Qiu Y, Yang X, Mao H, Huang F. Plasma fibrinogen and mortality in patients undergoing peritoneal dialysis: a prospective cohort study. BMC Nephrol 2020; 21:349. [PMID: 32807121 PMCID: PMC7430005 DOI: 10.1186/s12882-020-01984-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 07/27/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Plasma fibrinogen is significantly associated with cardiovascular (CV) events and mortality in the general population. However, the association between plasma fibrinogen and mortality in patients undergoing peritoneal dialysis (PD) is unclear. METHODS This was a prospective cohort study. A total of 1603 incident PD patients from a single center in South China were followed for a median of 46.7 months. A Cox regression analysis was used to evaluate the independent association of plasma fibrinogen with CV and all-cause mortality. Models were adjusted for age, sex, smoking, a history of CV events, diabetes, body mass index, systolic blood pressure, hemoglobin, blood platelet count, serum potassium, serum albumin, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, hypersensitive C-reactive protein, estimated glomerular filtration rate, antiplatelet agents and lipid-lowering drugs. RESULTS The mean age was 47.4 ± 15.3 years, 955 (59.6%) patients were male, 319 (19.9%) had a history of CV events, and 410 (25.6%) had diabetes. The average plasma fibrinogen level was 4.12 ± 1.38 g/L. Of the 474 (29.6%) patients who died during follow-up, 235 (49.6%) died due to CV events. In multivariable models, the adjusted hazard ratios (HRs) for quartile 1, quartile 3, and quartile 4 versus quartile 2 were 1.18 (95% confidence interval [CI], 0.72-1.95, P = 0.51), 1.47 (95% CI, 0.93-2.33, P = 0.10), and 1.78 (95% CI, 1.15-2.77, P = 0.01) for CV mortality and 1.20 (95% CI, 0.86-1.68, P = 0.28), 1.29 (95% CI, 0.93-1.78, P = 0.13), and 1.53 (95% CI, 1.12-2.09, P = 0.007) for all-cause mortality, respectively. A nonlinear relationship between plasma fibrinogen and CV and all-cause mortality was observed. CONCLUSIONS An elevated plasma fibrinogen level was significantly associated with an increased risk of CV and all-cause mortality in patients undergoing PD.
Collapse
Affiliation(s)
- Jing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Tong Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Naya Huang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xi Xia
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jianbo Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yagui Qiu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiao Yang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Fengxian Huang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China. .,Key Laboratory of Nephrology, National Health Commission and Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
12
|
Makhloufi C, Crescence L, Darbousset R, McKay N, Massy ZA, Dubois C, Panicot-Dubois L, Burtey S, Poitevin S. Assessment of Thrombotic and Bleeding Tendency in Two Mouse Models of Chronic Kidney Disease: Adenine-Diet and 5/6th Nephrectomy. TH OPEN 2020; 4:e66-e76. [PMID: 32309772 PMCID: PMC7162676 DOI: 10.1055/s-0040-1705138] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/27/2020] [Indexed: 12/15/2022] Open
Abstract
The coexistence of bleeding and thrombosis in patients with chronic kidney disease (CKD) is frequent and poorly understood. Mouse models are essential to understand complications of CKD and to develop new therapeutic approaches improving the health of patients. We evaluated the hemostasis in two models of renal insufficiency: adenine-diet and 5/6th nephrectomy (5/6Nx). Compared with 5/6Nx mice, mice fed with 0.25% adenine had more severe renal insufficiency and so higher levels of prothrombotic uremic toxins like indoxyl sulfate. More severe renal inflammation and fibrosis were observed in the adenine group, as demonstrated by histological and reverse transcription quantitative polymerase chain reaction experiments. Liver fibrinogen γ chain expression and level of plasma fibrinogen were increased only in adenine mice. In both CKD mouse models, tissue factor (TF) expression was increased in kidney and aorta extracts. Immunochemistry analysis of kidney sections showed that TF is localized in the vascular walls. Thrombin–antithrombin complexes were significantly increased in plasma from both adenine and 5/6Nx mice. Tail bleeding time increased significantly only in adenine mice, whereas platelet count was not significant altered. Finally, results obtained by intravital microscopy after laser-induced endothelial injury showed impaired platelet function in adenine mice and an increase in fibrin generation in 5/6Nx mice. To summarize, adenine diet causes a more severe renal insufficiency compared with 5/6Nx. The TF upregulation and the hypercoagulable state were observed in both CKD models. Bleeding tendency was observed only in the adenine model of CKD that recapitulates the whole spectrum of hemostasis abnormalities observed in advanced human CKD.
Collapse
Affiliation(s)
| | - Lydie Crescence
- Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
| | - Roxane Darbousset
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Nathalie McKay
- Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
| | - Ziad A Massy
- Centre for Research in Epidemiology and Population Health (CESP), University Paris-Saclay, Villejuif, France.,Department of Nephrology, Ambroise Paré University Hospital, Boulogne Billancourt/Paris, France
| | | | | | - Stéphane Burtey
- Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France.,Centre de Néphrologie et Transplantation Rénale, APHM, Marseille, France
| | | |
Collapse
|
13
|
Nattokinase-heparin exhibits beneficial efficacy and safety-an optimal strategy for CKD patients on hemodialysis. Glycoconj J 2019; 36:93-101. [PMID: 30788657 DOI: 10.1007/s10719-019-09860-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/20/2018] [Accepted: 01/24/2019] [Indexed: 12/19/2022]
Abstract
Heparin is a widely used anticoagulant in hemodialysis (HD) for patients with chronic kidney disease (CKD); however, it entails the risk of thrombus formation due to heparin-induced thrombocytopenia. Indeed, CKD patients on HD are associated with excessive mortality from cardiovascular disease due to their prothrombotic profile. Therefore, it would be a significant breakthrough to develop a thrombolytic adjuvant that facilitates heparin to achieve its proper anticoagulant efficiency at a much lower dose for greater safety. Nattokinase (NK), a valuable dietary supplement possessing strong fibrinolytic and thrombolytic activity, was reported to interact with heparin and thereby the beneficial efficacy of NK-heparin was investigated herein. NK-heparin induced a synergistic enhancement of clotting time both in vitro and in vivo evaluations, whereas the overall fibrinolytic activity was only marginally enhanced. Moreover, it was demonstrated for the first time that NK induced potent degradation of all three chains of fibrinogen. In particular, NK-heparin markedly reinforced the fibrinolysis activity of NK, which may underlie, at least in part, the mechanism by which NK-heparin benefited their overall thrombolytic and anticoagulant activity. Collectively, we clarified the beneficial combination efficacy of NK and heparin for greater safety, providing a powerful impetus for physicians to administer heparin to a larger portion of patients with CKD.
Collapse
|
14
|
Blood loss mitigation and replacement in facial surgery: a review. Curr Opin Otolaryngol Head Neck Surg 2018; 26:266-274. [PMID: 29846240 DOI: 10.1097/moo.0000000000000461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW To provide a comprehensive overview of the predisposing factors that contribute to a risk of excess bleeding for surgical therapy in the head and neck regions, provide a thorough overview of techniques and tools for managing blood loss complications, and provide intervention algorithms to help guide clinical decision making. RECENT FINDINGS With the current landscape of medications and reversal agents, protocols for intervention in a variety of situations, and new tools for blood loss management all rapidly changing and being developed it is critical to stay up to date to provide patients the best care in the most critical of situations. SUMMARY With the risk of blood loss complications in head and neck surgery ranging from minimal to extreme surgeons require a comprehensive understanding risk factors, patient evaluation tools, and proper management algorithms. The first opportunity to prevent unnecessary blood loss and blood loss complications is the health history and physical appointment where a clinician can identify any medications, conditions, or other predisposing factors that would elevate a patient's risk of excess bleeding and the necessity for treatment augmentation. Although not all complications can be prevented because of the natural physiological variation that occurs from patient to patient, despite proper and proper diagnostics, a full working knowledge of most likely complications, hemostatic tools, and concise communication with team members can prevent a lot of blood loss and the complications associated.
Collapse
|
15
|
Addi T, Dou L, Burtey S. Tryptophan-Derived Uremic Toxins and Thrombosis in Chronic Kidney Disease. Toxins (Basel) 2018; 10:E412. [PMID: 30322010 PMCID: PMC6215213 DOI: 10.3390/toxins10100412] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Patients with chronic kidney disease (CKD) display an elevated risk of thrombosis. Thrombosis occurs in cardiovascular events, such as venous thromboembolism, stroke, and acute coronary syndrome, and is a cause of hemodialysis vascular access dysfunction. CKD leads to the accumulation of uremic toxins, which exerts toxic effects on blood and the vessel wall. Some uremic toxins result from tryptophan metabolization in the gut through the indolic and the kynurenine pathways. An increasing number of studies are highlighting the link between such uremic toxins and thrombosis in CKD. In this review, we describe the thrombotic mechanisms induced by tryptophan-derived uremic toxins (TDUT). These mechanisms include an increase in plasma levels of procoagulant factors, induction of platelet hyperactivity, induction of endothelial dysfunction/ impairment of endothelial healing, decrease in nitric oxide (NO) bioavailability, and production of procoagulant microparticles. We focus on one important prothrombotic mechanism: The induction of tissue factor (TF), the initiator of the extrinsic pathway of the blood coagulation. This induction occurs via a new pathway, dependent on the transcription factor Aryl hydrocarbon receptor (AhR), the receptor of TDUT in cells. A better understanding of the prothrombotic mechanisms of uremic toxins could help to find novel therapeutic targets to prevent thrombosis in CKD.
Collapse
Affiliation(s)
- Tawfik Addi
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France.
- LPNSA, Département de Biologie, Université d'Oran 1 Ahmed Benbella, 31000 Oran, Algérie.
| | - Laetitia Dou
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France.
| | - Stéphane Burtey
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France.
- Centre de Néphrologie et Transplantation Rénale, AP-HM, 13005 Marseille, France.
| |
Collapse
|
16
|
Bonello L, Angiolillo DJ, Aradi D, Sibbing D. P2Y
12
-ADP Receptor Blockade in Chronic Kidney Disease Patients With Acute Coronary Syndromes. Circulation 2018; 138:1582-1596. [DOI: 10.1161/circulationaha.118.032078] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Laurent Bonello
- Aix-Marseille Université, INSERM UMR-S 1076, Vascular Research Center of Marseille, Marseille, France (L.B.)
| | - Dominick J. Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville (D.J.A.)
| | - Daniel Aradi
- Heart Center Balatonfüred and Semmelweis University Budapest, Hungary (D.A.)
| | - Dirk Sibbing
- Department of Cardiology, Ludwig-Maximilians-Universität München, Germany (D.S.)
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.S.)
| |
Collapse
|
17
|
Watanabe Y, Koyama S, Yamashita A, Matsuura Y, Nishihira K, Kitamura K, Asada Y. Indoleamine 2,3-dioxygenase 1 in coronary atherosclerotic plaque enhances tissue factor expression in activated macrophages. Res Pract Thromb Haemost 2018; 2:726-735. [PMID: 30349892 PMCID: PMC6178752 DOI: 10.1002/rth2.12128] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 06/04/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent clinical studies have found that changes in the kynurenine (Kyn) pathway of tryptophan (Trp) metabolism are associated with cardiovascular events. However, the roles of the Kyn pathway on vascular wall thrombogenicity remain unknown. Indoleamine 2,3-dioxygenase 1 (IDO1) is a rate-limiting enzyme of the Kyn pathway. OBJECTIVE The present study aimed to localize IDO1 in human coronary atherosclerotic plaques from patients with angina pectoris and define its role in plaque thrombogenicity. METHODS Immunohistochemical methods were applied to localize IDO1 in coronary atherosclerotic plaques from patients with stable (SAP) and unstable (UAP) angina pectoris. The role of IDO1 in tissue factor (TF) expression was investigated in THP-1 macrophages activated by interferon (IFN)γ and tissue necrosis factor (TNF)α. RESULTS We localized IDO1 mainly in CD68-positive macrophages within atherosclerotic plaques, and in close association with TF. Areas that were immunopositive for IDO1, TF, and CD3-positive T lymphocytes were significantly larger in plaques from patients with UAP than SAP. Macrophages activated by IFNγ and TNFα upregulated IDO1 expression, increased the Kyn/Trp ratio and enhanced TF expression and activity, but not TF pathway inhibitor expression. The IDO1 inhibitor epacadostat significantly reduced the Kyn/Trp ratio, TF expression and activity, as well as NF-κB (p65) binding activity in activated macrophages. Inhibition of the aryl hydrocarbon receptor that binds to Kyn, also reduced Kyn-induced TF expression in activated macrophages. CONCLUSION Indoleamine 2,3-dioxygenase 1 expressed in coronary atherosclerotic plaques might contribute to thrombus formation through TF upregulation in activated macrophages.
Collapse
Affiliation(s)
- Yuki Watanabe
- Department of PathologyFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
- Department of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Shohei Koyama
- Department of PathologyFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
- Department of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Atsushi Yamashita
- Department of PathologyFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Yunosuke Matsuura
- Department of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Kensaku Nishihira
- Department of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Kazuo Kitamura
- Department of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Yujiro Asada
- Department of PathologyFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| |
Collapse
|
18
|
Song TJ, Kwon I, Piao H, Lee JE, Han KR, Chang Y, Oh HJ, Choi HJ, Lee KY, Kim YJ, Han KH, Heo JH. Increased Thrombogenicity in Chronic Renal Failure in a Rat Model Induced by 5/6 Ablation/Infarction. Yonsei Med J 2018; 59:754-759. [PMID: 29978612 PMCID: PMC6037604 DOI: 10.3349/ymj.2018.59.6.754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/25/2018] [Accepted: 05/23/2018] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Abnormalities in hemostasis and coagulation have been suggested in chronic renal failure (CRF). In this study, we compared processes of thrombus formation between rats with CRF and those with normal kidney function. MATERIALS AND METHODS CRF was induced by 5/6 ablation/infarction of the kidneys in Sprague-Dawley rats, and surviving rats after 4 weeks were used. Ferric chloride (FeCl₃)-induced thrombosis in the carotid artery was induced to assess thrombus formation. Whole blood clot formation was evaluated using rotational thromboelastometry (ROTEM). Platelet aggregation was assessed with impedance platelet aggregometry. RESULTS FeCl₃-induced thrombus formation was initiated faster in the CRF group than in the control group (13.2±1.1 sec vs. 17.8±1.0 sec, p=0.027). On histological examination, the maximal diameters of thrombi were larger in the CRF group than in the control group (394.2±201.1 μm vs. 114.0±145.1 μm, p=0.039). In extrinsic pathway ROTEM, the CRF group showed faster clot initiation (clotting time, 59.0±7.3 sec vs. 72.8±5.0 sec, p=0.032) and increased clot growth kinetics (α angle, 84.8±0.2° vs. 82.0±0.6°, p=0.008), compared to the control group. Maximal platelet aggregation rate was higher in the CRF group than in the control group (58.2±0.2% vs. 44.6±1.2%, p=0.006). CONCLUSION Our study demonstrated that thrombogenicity is increased in rats with CRF. An activated extrinsic coagulation pathway may play an important role in increasing thrombogenicity in CRF.
Collapse
Affiliation(s)
- Tae Jin Song
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Korea
| | - Il Kwon
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Honglim Piao
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, Korea
| | - Jee Eun Lee
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Korea
| | - Kyeo Rye Han
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Korea
| | - Yoonkyung Chang
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Korea
| | - Hyung Jung Oh
- Ewha Institute of Convergence Medicine, Ewha Womans University, Seoul, Korea
| | - Hyun Jung Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Yul Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Jae Kim
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Korea
| | - Ki Hwan Han
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, Korea
| | - Ji Hoe Heo
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
19
|
Kolachalama VB, Shashar M, Alousi F, Shivanna S, Rijal K, Belghasem ME, Walker J, Matsuura S, Chang GH, Gibson CM, Dember LM, Francis JM, Ravid K, Chitalia VC. Uremic Solute-Aryl Hydrocarbon Receptor-Tissue Factor Axis Associates with Thrombosis after Vascular Injury in Humans. J Am Soc Nephrol 2018; 29:1063-1072. [PMID: 29343519 DOI: 10.1681/asn.2017080929] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/21/2017] [Indexed: 11/03/2022] Open
Abstract
Individuals with CKD are particularly predisposed to thrombosis after vascular injury. Using mouse models, we recently described indoxyl sulfate, a tryptophan metabolite retained in CKD and an activator of tissue factor (TF) through aryl hydrocarbon receptor (AHR) signaling, as an inducer of thrombosis across the CKD spectrum. However, the translation of findings from animal models to humans is often challenging. Here, we investigated the uremic solute-AHR-TF thrombosis axis in two human cohorts, using a targeted metabolomics approach to probe a set of tryptophan products and high-throughput assays to measure AHR and TF activity. Analysis of baseline serum samples was performed from 473 participants with advanced CKD from the Dialysis Access Consortium Clopidogrel Prevention of Early AV Fistula Thrombosis trial. Participants with subsequent arteriovenous thrombosis had significantly higher levels of indoxyl sulfate and kynurenine, another uremic solute, and greater activity of AHR and TF, than those without thrombosis. Pattern recognition analysis using the components of the thrombosis axis facilitated clustering of the thrombotic and nonthrombotic groups. We further validated these findings using 377 baseline samples from participants in the Thrombolysis in Myocardial Infarction II trial, many of whom had CKD stage 2-3. Mechanistic probing revealed that kynurenine enhances thrombosis after vascular injury in an animal model and regulates thrombosis in an AHR-dependent manner. This human validation of the solute-AHR-TF axis supports further studies probing its utility in risk stratification of patients with CKD and exploring its role in other diseases with heightened risk of thrombosis.
Collapse
Affiliation(s)
- Vijaya B Kolachalama
- Section of Computational Biomedicine and.,Department of Medicine, Whitaker Cardiovascular Institute, and.,Hariri Institute for Computing and Computational Science and Engineering, Boston University, Boston, Massachusetts
| | | | | | | | | | - Mostafa E Belghasem
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| | | | | | | | - C Michael Gibson
- Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; and
| | - Laura M Dember
- Renal-Electrolyte and Hypertension Division, Department of Medicine, Center for Clinical Epidemiology and Biostatistics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Katya Ravid
- Department of Medicine, Whitaker Cardiovascular Institute, and
| | - Vipul C Chitalia
- Department of Medicine, Whitaker Cardiovascular Institute, and .,Renal Section, Department of Medicine.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
20
|
Haghighi AG, Finder RG, Bennett JD. Systemic Disease and Bleeding Disorders for the Oral and Maxillofacial Surgeon. Oral Maxillofac Surg Clin North Am 2017; 28:461-471. [PMID: 27745617 DOI: 10.1016/j.coms.2016.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There are multiple systemic diseases that have an impact on coagulation, of which oral and maxillofacial surgeons must be cognizant. Recent evidence has supported the potential for both hypocoagulable and hypercoagulable states in patients with liver and kidney disease with an even less understood impact on prolonged bleeding in the oral cavity. These systemic diseases are not limited to diseases affecting the liver, kidney, and bone marrow; however, these diseases are common among the patient population and surgeons must be capable of making appropriate judgment and modifying care appropriately.
Collapse
Affiliation(s)
- Arman G Haghighi
- Department of Oral and Maxillofacial Surgery, Indiana University, Indianapolis, IN, USA.
| | - Rebecca G Finder
- Critical Care Pharmacy, Indiana University Health, 1701 N. Senate Blvd, Indianapolis, IN 46202, USA
| | - Jeffrey D Bennett
- Division of Oral and Maxillofacial Surgery, Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
21
|
Rysz J, Gluba-Brzózka A, Franczyk B, Jabłonowski Z, Ciałkowska-Rysz A. Novel Biomarkers in the Diagnosis of Chronic Kidney Disease and the Prediction of Its Outcome. Int J Mol Sci 2017; 18:E1702. [PMID: 28777303 PMCID: PMC5578092 DOI: 10.3390/ijms18081702] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/17/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
In its early stages, symptoms of chronic kidney disease (CKD) are usually not apparent. Significant reduction of the kidney function is the first obvious sign of disease. If diagnosed early (stages 1 to 3), the progression of CKD can be altered and complications reduced. In stages 4 and 5 extensive kidney damage is observed, which usually results in end-stage renal failure. Currently, the diagnosis of CKD is made usually on the levels of blood urea and serum creatinine (sCr), however, sCr has been shown to be lacking high predictive value. Due to the development of genomics, epigenetics, transcriptomics, proteomics, and metabolomics, the introduction of novel techniques will allow for the identification of novel biomarkers in renal diseases. This review presents some new possible biomarkers in the diagnosis of CKD and in the prediction of outcome, including asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA), uromodulin, kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), miRNA, ncRNA, and lincRNA biomarkers and proteomic and metabolomic biomarkers. Complicated pathomechanisms of CKD development and progression require not a single marker but their combination in order to mirror all types of alterations occurring in the course of this disease. It seems that in the not so distant future, conventional markers may be exchanged for new ones, however, confirmation of their efficacy, sensitivity and specificity as well as the reduction of analysis costs are required.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, WAM Teaching Hospital, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Zbigniew Jabłonowski
- I Department of Urology, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Aleksandra Ciałkowska-Rysz
- Palliative Medicine Unit, Chair of Oncology, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| |
Collapse
|
22
|
Schuett K, Savvaidis A, Maxeiner S, Lysaja K, Jankowski V, Schirmer SH, Dimkovic N, Boor P, Kaesler N, Dekker FW, Floege J, Marx N, Schlieper G. Clot Structure: A Potent Mortality Risk Factor in Patients on Hemodialysis. J Am Soc Nephrol 2017; 28:1622-1630. [PMID: 28057772 DOI: 10.1681/asn.2016030336] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 10/22/2016] [Indexed: 11/03/2022] Open
Abstract
Patients with CKD on hemodialysis exhibit increased cardiovascular risk. Fibrin clot structure and clot lysis are crucially involved in development of cardiovascular events, but little is known about the influence of clot density on outcome in patients on hemodialysis. We determined fibrin clot structure parameters and effect on mortality in a prospective cohort of 171 patients on chronic hemodialysis (mean±SD age =59±11 years old; 54% men) using a validated turbidimetric assay. Kaplan-Meier analysis revealed that patients on hemodialysis with a denser clot structure had increased all-cause and cardiovascular mortality risks (log rank P=0.004 and P=0.003, respectively). Multivariate Cox regression models (adjusted for age, diabetes, sex, and duration of dialysis or fibrinogen, C-reactive protein, and complement C3) confirmed that denser clots are independently related to mortality risk. We also purified fibrinogen from healthy controls and patients on hemodialysis using the calcium-dependent IF-1 mAb against fibrinogen for additional investigation using mass spectrometric analysis and electron microscopy. Whereas purified fibrinogen from healthy controls displayed no post-translational modifications, fibrinogen from patients on hemodialysis was glycosylated and guanidinylated. Clots made of purified fibrinogen from patients on hemodialysis exhibited significantly thinner fibers compared with clots from fibrinogen of control individuals (mean±SD =63±2 and 77±2 nm, respectively; P<0.001). In vitro guanidinylation of fibrinogen from healthy subjects increased the formation of thinner fibers, suggesting that difference in fiber thickness might be at least partially due to post-translational modifications. Thus, in patients on hemodialysis, a denser clot structure may be a potent independent risk factor for mortality.
Collapse
Affiliation(s)
| | | | | | | | - Vera Jankowski
- Institute for Molecular Cardiovascular Research, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Stephan H Schirmer
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Nada Dimkovic
- Center for Renal Diseases, Zvezdara University Medical Center, Belgrade, Serbia
| | | | | | - Friedo W Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands; and
| | | | | | - Georg Schlieper
- Internal Medicine II and.,Medizinisches Versorgungszentrum DaVita Rhein-Ruhr, Düsseldorf, Germany
| |
Collapse
|
23
|
Shen K, Johnson DW, Gobe GC. The role of cGMP and its signaling pathways in kidney disease. Am J Physiol Renal Physiol 2016; 311:F671-F681. [PMID: 27413196 DOI: 10.1152/ajprenal.00042.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/10/2016] [Indexed: 01/20/2023] Open
Abstract
Cyclic nucleotide signal transduction pathways are an emerging research field in kidney disease. Activated cell surface receptors transduce their signals via intracellular second messengers such as cAMP and cGMP. There is increasing evidence that regulation of the cGMP-cGMP-dependent protein kinase 1-phosphodiesterase (cGMP-cGK1-PDE) signaling pathway may be renoprotective. Selective PDE5 inhibitors have shown potential in treating kidney fibrosis in patients with chronic kidney disease (CKD), via their downstream signaling, and these inhibitors also have known activity as antithrombotic and anticancer agents. This review gives an outline of the cGMP-cGK1-PDE signaling pathways and details the downstream signaling and regulatory functions that are modulated by cGK1 and PDE inhibitors with regard to antifibrotic, antithrombotic, and antitumor activity. Current evidence that supports the renoprotective effects of regulating cGMP-cGK1-PDE signaling is also summarized. Finally, the effects of icariin, a natural plant extract with PDE5 inhibitory function, are discussed. We conclude that regulation of cGMP-cGK1-PDE signaling might provide novel, therapeutic strategies for the worsening global public health problem of CKD.
Collapse
Affiliation(s)
- Kunyu Shen
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China; and
| | - David W Johnson
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia;
| |
Collapse
|
24
|
Chang YS, Weng SF, Chang C, Wang JJ, Tseng SH, Wang JY, Jan RL. Risk of Retinal Vein Occlusion Following End-Stage Renal Disease. Medicine (Baltimore) 2016; 95:e3474. [PMID: 27100450 PMCID: PMC4845854 DOI: 10.1097/md.0000000000003474] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The aim of the study was to investigate the risk of retinal vein occlusion (RVO) following end-stage renal disease (ESRD). The study was designed as a retrospective, nationwide, matched cohort study. The subjects were ESRD patients identified by the International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM), code 585. The study cohort included 92,774 ESRD patients registered between January 2000 and December 2009 at the Taiwan National Health Insurance Research Database. An age- and sex-matched control group comprised 92,774 patients (case:control = 1:1) selected from the Taiwan Longitudinal Health Insurance Database 2000. Information for each patient was collected from the index date until December 2011. The incidence and risk of RVO were compared between the ESRD and control groups. The adjusted hazard ratio (HR) for RVO after adjustment for potential confounders was obtained by Cox proportional hazard regression analysis. Kaplan-Meier analysis was used to calculate the RVO cumulative incidence rate. The main outcome measure was the incidence of RVO following ESRD. In total, 904 ESRD patients (0.97%) and 410 controls (0.44%) had RVO (P < 0.0001) during the follow-up period, leading to a significantly elevated risk of RVO in the ESRD patients compared with controls (incidence rate ratio = 3.05, 95% confidence interval = 2.72-3.43). After adjustment for potential confounders including diabetes mellitus, hypertension, hyperlipidemia, congestive heart failure, and coronary artery disease, ESRD patients were 3.05 times more likely to develop RVO in the full cohort (adjusted hazard ratio = 3.05, 95% confidence interval = 2.64-3.51). In addition, hypertension patients showed high incidence rate of RVO in the ESRD group compared with controls (incidence rate ratio = 1.71, 95% confidence interval = 1.44-2.03) and maintained significant risk of RVO after adjustment for other confounders in the cohort (adjusted hazard ratio = 1.39, 95% confidence interval = 1.20-1.60). ESRD increases the risk of RVO. For ESRD patients, we recommend education regarding RVO in addition to blood pressure control to prevent subsequent RVO.
Collapse
Affiliation(s)
- Yuh-Shin Chang
- From the Department of Ophthalmology (Y-SC, S-HT), Chi Mei Medical Center, Tainan, Taiwan; Graduate Institute of Medical Science (Y-SC), College of Health Science, Chang Jung Christian University, Tainan, Taiwan; Department of Healthcare Administration and Medical Informatics (S-FW), Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Education (CC), University of Taipei, Taipei, Taiwan; Department of Anesthesiology (J-JW), Chi Mei Medical Center, Tainan, Taiwan; Department of Ophthalmology (S-HT), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Graduate Institute of Clinical Medicine (J-YW, R-LJ), National Cheng Kung University, Tainan, Taiwan; and Department of Pediatrics (R-LJ), Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
25
|
Schlieper G, Hess K, Floege J, Marx N. The vulnerable patient with chronic kidney disease. Nephrol Dial Transplant 2015; 31:382-90. [DOI: 10.1093/ndt/gfv041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 01/25/2015] [Indexed: 11/14/2022] Open
|
26
|
Nkuipou-Kenfack E, Duranton F, Gayrard N, Argilés À, Lundin U, Weinberger KM, Dakna M, Delles C, Mullen W, Husi H, Klein J, Koeck T, Zürbig P, Mischak H. Assessment of metabolomic and proteomic biomarkers in detection and prognosis of progression of renal function in chronic kidney disease. PLoS One 2014; 9:e96955. [PMID: 24817014 PMCID: PMC4016198 DOI: 10.1371/journal.pone.0096955] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/14/2014] [Indexed: 11/18/2022] Open
Abstract
Chronic kidney disease (CKD) is part of a number of systemic and renal diseases and may reach epidemic proportions over the next decade. Efforts have been made to improve diagnosis and management of CKD. We hypothesised that combining metabolomic and proteomic approaches could generate a more systemic and complete view of the disease mechanisms. To test this approach, we examined samples from a cohort of 49 patients representing different stages of CKD. Urine samples were analysed for proteomic changes using capillary electrophoresis-mass spectrometry and urine and plasma samples for metabolomic changes using different mass spectrometry-based techniques. The training set included 20 CKD patients selected according to their estimated glomerular filtration rate (eGFR) at mild (59.9±16.5 mL/min/1.73 m2; n = 10) or advanced (8.9±4.5 mL/min/1.73 m2; n = 10) CKD and the remaining 29 patients left for the test set. We identified a panel of 76 statistically significant metabolites and peptides that correlated with CKD in the training set. We combined these biomarkers in different classifiers and then performed correlation analyses with eGFR at baseline and follow-up after 2.8±0.8 years in the test set. A solely plasma metabolite biomarker-based classifier significantly correlated with the loss of kidney function in the test set at baseline and follow-up (ρ = -0.8031; p<0.0001 and ρ = -0.6009; p = 0.0019, respectively). Similarly, a urinary metabolite biomarker-based classifier did reveal significant association to kidney function (ρ = -0.6557; p = 0.0001 and ρ = -0.6574; p = 0.0005). A classifier utilising 46 identified urinary peptide biomarkers performed statistically equivalent to the urinary and plasma metabolite classifier (ρ = -0.7752; p<0.0001 and ρ = -0.8400; p<0.0001). The combination of both urinary proteomic and urinary and plasma metabolic biomarkers did not improve the correlation with eGFR. In conclusion, we found excellent association of plasma and urinary metabolites and urinary peptides with kidney function, and disease progression, but no added value in combining the different biomarkers data.
Collapse
Affiliation(s)
- Esther Nkuipou-Kenfack
- Mosaiques Diagnostics GmbH, Hannover, Germany
- Department of Toxicology, Hannover Medical School, Hannover, Germany
| | | | | | - Àngel Argilés
- RD Néphrologie, Montpellier, France
- Néphrologie Dialyse St Guilhem, Séte, France
- Service de Néphrologie, Dialyse Péritonéale et Transplantation, Montpellier, France
| | | | - Klaus M. Weinberger
- Biocrates life sciences AG, Innsbruck, Austria
- sAnalytiCo Ltd, Belfast, United Kingdom
- Department of Biomedical Informatics and Mechatronics, Private University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
| | | | - Christian Delles
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - William Mullen
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Holger Husi
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Julie Klein
- Mosaiques Diagnostics GmbH, Hannover, Germany
| | | | | | - Harald Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
Lutz J, Menke J, Sollinger D, Schinzel H, Thürmel K. Haemostasis in chronic kidney disease. Nephrol Dial Transplant 2013; 29:29-40. [PMID: 24132242 DOI: 10.1093/ndt/gft209] [Citation(s) in RCA: 276] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The coagulation system has gained much interest again as new anticoagulatory substances have been introduced into clinical practice. Especially patients with renal failure are likely candidates for such a therapy as they often experience significant comorbidity including cardiovascular diseases that require anticoagulation. Patients with renal failure on new anticoagulants have experienced excessive bleeding which can be related to a changed pharmacokinetic profile of the compounds. However, the coagulation system itself, even without any interference with coagulation modifying drugs, is already profoundly changed during renal failure. Coagulation disorders with either episodes of severe bleeding or thrombosis represent an important cause for the morbidity and mortality of such patients. The underlying reasons for these coagulation disorders involve the changed interaction of different components of the coagulation system such as the coagulation cascade, the platelets and the vessel wall in the metabolic conditions of renal failure. Recent work provides evidence that new factors such as microparticles (MPs) can influence the coagulation system in patients with renal insufficiency through their potent procoagulatory effects. Interestingly, MPs may also contain microRNAs thus inhibiting the function of platelets, resulting in bleeding episodes. This review comprises the findings on the complex pathophysiology of coagulation disorders including new factors such as MPs and microRNAs in patients with renal insufficiency.
Collapse
Affiliation(s)
- Jens Lutz
- Schwerpunkt Nephrologie, I. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes Gutenberg Universität Mainz, Mainz, Germany
| | | | | | | | | |
Collapse
|
28
|
Coagulation and coagulation signalling in fibrosis. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1018-27. [PMID: 23298546 DOI: 10.1016/j.bbadis.2012.12.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 12/20/2012] [Accepted: 12/22/2012] [Indexed: 12/29/2022]
Abstract
Following tissue injury, a complex and coordinated wound healing response comprising coagulation, inflammation, fibroproliferation and tissue remodelling has evolved to nullify the impact of the original insult and reinstate the normal physiological function of the affected organ. Tissue fibrosis is thought to result from a dysregulated wound healing response as a result of continual local injury or impaired control mechanisms. Although the initial insult is highly variable for different organs, in most cases, uncontrolled or sustained activation of mesenchymal cells into highly synthetic myofibroblasts leads to the excessive deposition of extracellular matrix proteins and eventually loss of tissue function. Coagulation was originally thought to be an acute and transient response to tissue injury, responsible primarily for promoting haemostasis by initiating the formation of fibrin plugs to enmesh activated platelets within the walls of damaged blood vessels. However, the last 20years has seen a major re-evaluation of the role of the coagulation cascade following tissue injury and there is now mounting evidence that coagulation plays a critical role in orchestrating subsequent inflammatory and fibroproliferative responses during normal wound healing, as well as in a range of pathological contexts across all major organ systems. This review summarises our current understanding of the role of coagulation and coagulation initiated signalling in the response to tissue injury, as well as the contribution of uncontrolled coagulation to fibrosis of the lung, liver, kidney and heart. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
|
29
|
Pawlak K, Ulazka B, Mysliwiec M, Pawlak D. Vascular endothelial growth factor and uPA/suPAR system in early and advanced chronic kidney disease patients: a new link between angiogenesis and hyperfibrinolysis? Transl Res 2012; 160:346-54. [PMID: 22683425 DOI: 10.1016/j.trsl.2012.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 04/04/2012] [Accepted: 04/12/2012] [Indexed: 10/28/2022]
Abstract
Disturbances in hemostasis and abnormal angiogenesis are components in the plaque growth and destabilization. The role of the vascular endothelial growth factor (VEGF) in the perturbation of hemostasis in chronic kidney disease (CKD) is still unknown. In this preliminary study, we investigate the relation between VEGF and the parameters of coagulation: tissue factor (TF), its inhibitor (TFPI), and fibrinolytic system: urokinase-type plasminogen activator (uPA), its soluble receptor (suPAR), tissue-type plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1), plasmin/antiplasmin complexes (PAP) in the patients with mild-to-moderate, and severe CKD and healthy controls. All indices (except TFPI) were raised in CKD patients, particularly in those with severe CKD, compared with controls. The strong positive correlations were between VEGF and some parameters, both coagulation (TF, TFPI, TF/TFPI ratio) and fibrinolytic system (uPA, suPAR, PAP). The relationships were also between the individual hemostatic parameters. In multiple regression analysis, VEGF and kidney dysfunction markers (urea and creatinine levels) were independently associated with uPA, and VEGF was independently associated with suPAR levels. Moreover, PAP was independently associated with age and suPAR. This study represents the first to investigate the relation between VEGF and the activation both coagulation and fibrinolysis in CKD patients. VEGF and the parameters of hemostatic system activation were higher in the CKD group than in the controls with a significant correlation between them. VEGF was independently associated with uPA/suPAR system, whereas suPAR was independently related to PAP levels, suggesting a new link between abnormal angiogenesis and hyperfibrinolysis in this population.
Collapse
Affiliation(s)
- Krystyna Pawlak
- Department of Monitored Pharmacotherapy, Medical University, Bialystok, Poland.
| | | | | | | |
Collapse
|
30
|
Kumar G, Sakhuja A, Taneja A, Majumdar T, Patel J, Whittle J, Nanchal R. Pulmonary embolism in patients with CKD and ESRD. Clin J Am Soc Nephrol 2012; 7:1584-90. [PMID: 22837271 DOI: 10.2215/cjn.00250112] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES CKD and ESRD are growing burdens. It is unclear whether these conditions affect pulmonary embolism (PE) risk, given that they affect both procoagulant and anticoagulant factors. This study examined the frequency and associated outcomes of PE in CKD and ESRD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS The Healthcare Cost and Utilization Project's Nationwide Inpatient Sample was used to estimate the frequency and outcomes of PE in adults with CKD and ESRD. Hospitalizations for the principal diagnosis of PE and presence of CKD or ESRD were identified using International Classification of Diseases, Ninth Revision codes. Data from the annual US Census and US Renal Data System reports were used to calculate the number of adults with CKD, ESRD, and normal kidney function (NKF) as well as the annual incidence of PE in each group. Logistic regression modeling was used to compare in-hospital mortality among persons admitted for PE who had ESRD or CKD to those without these conditions. RESULTS The annual frequency of PE was 527 per 100,000, 204 per 100,000, and 66 per 100,000 persons with ESRD, CKD, and NKF, respectively. In-hospital mortality was higher for persons with ESRD and CKD (P<0.001) compared with persons with NKF. Median length of stay was longer by 1 day in CKD and 2 days in ESRD than among those with NKF. CONCLUSIONS Persons with CKD and ESRD are more likely to have PE than persons with NKF. Once they have PE, they are more likely to die in the hospital.
Collapse
Affiliation(s)
- Gagan Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Shing CM, Adams MJ, Fassett RG, Coombes JS. Nutritional compounds influence tissue factor expression and inflammation of chronic kidney disease patients in vitro. Nutrition 2011; 27:967-72. [PMID: 21295946 DOI: 10.1016/j.nut.2010.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 08/17/2010] [Accepted: 10/06/2010] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Nutritional compounds that potentially limit inflammation and tissue factor expression may decrease the progression of chronic kidney disease (CKD) and associated cardiovascular disease. This project aimed to determine the effect of curcumin, bovine colostrum, and fish oil on inflammatory cytokine and tissue factor procoagulant activity of peripheral blood mononuclear cells (PBMCs) from patients with CKD before dialysis. METHODS Peripheral blood mononuclear cells from patients with CKD before dialysis (n = 13) and age- and sex-matched healthy controls (n = 12) were cultured alone and with low and high doses of the nutritional compounds for 24 h. Cells were cultured with and without lipopolysaccharide. Supernatants were analyzed for tumor necrosis factor-α, interleukin (IL)-6, IL-8, monocyte chemoattractant protein-1, IL-1β, C-reactive protein, and tissue factor procoagulant activity. RESULTS The production of C-reactive protein, monocyte chemoattractant protein-1, IL-6, and IL-1β by PBMCs was inhibited by low- and high-dose fish oil in the CKD group (P < 0.05). Curcumin decreased secretion of IL-6 (P = 0.015) and IL-1 β (P = 0.016). Curcumin was more effective than colostrum at decreasing the procoagulant activity of PBMCs in the CKD and control groups (P < 0.019). CONCLUSION Fish oil decreased inflammatory cytokine secretion from CKD PBMCs. In addition, the beneficial effects of curcumin were demonstrated in decreasing inflammation in vitro, often to a similar magnitude as fish oil.
Collapse
Affiliation(s)
- Cecilia M Shing
- School of Human Life Sciences, University of Tasmania, Launceston, Australia.
| | | | | | | |
Collapse
|