Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Bioact Mater ; 38: 499-511, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38798890

ABSTRACT

The timely establishment of functional neo-vasculature is pivotal for successful tissue development and regeneration, remaining a central challenge in tissue engineering. In this study, we present a novel (micro)vascularization strategy that explores the use of specialized "vascular units" (VUs) as building blocks to initiate blood vessel formation and create perfusable, stroma-embedded 3D microvascular networks from the bottom-up. We demonstrate that VUs composed of endothelial progenitor cells and organ-specific fibroblasts exhibit high angiogenic potential when embedded in fibrin hydrogels. This leads to the formation of VUs-derived capillaries, which fuse with adjacent capillaries to form stable microvascular beds within a supportive, extracellular matrix-rich fibroblastic microenvironment. Using a custom-designed biomimetic fibrin-based vessel-on-chip (VoC), we show that VUs-derived capillaries can inosculate with endothelialized microfluidic channels in the VoC and become perfused. Moreover, VUs can establish capillary bridges between channels, extending the microvascular network throughout the entire device. When VUs and intestinal organoids (IOs) are combined within the VoC, the VUs-derived capillaries and the intestinal fibroblasts progressively reach and envelop the IOs. This promotes the formation of a supportive vascularized stroma around multiple IOs in a single device. These findings underscore the remarkable potential of VUs as building blocks for engineering microvascular networks, with versatile applications spanning from regenerative medicine to advanced in vitro models.

2.
Arch Toxicol ; 98(3): 929-942, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38197913

ABSTRACT

Adverse outcome pathways (AOPs) were introduced in modern toxicology to provide evidence-based representations of the events and processes involved in the progression of toxicological effects across varying levels of the biological organisation to better facilitate the safety assessment of chemicals. AOPs offer an opportunity to address knowledge gaps and help to identify novel therapeutic targets. They also aid in the selection and development of existing and new in vitro and in silico test methods for hazard identification and risk assessment of chemical compounds. However, many toxicological processes are too intricate to be captured in a single, linear AOP. As a result, AOP networks have been developed to aid in the comprehension and placement of associated events underlying the emergence of related forms of toxicity-where complex exposure scenarios and interactions may influence the ultimate adverse outcome. This study utilised established criteria to develop an AOP network that connects thirteen individual AOPs associated with nephrotoxicity (as sourced from the AOP-Wiki) to identify several key events (KEs) linked to various adverse outcomes, including kidney failure and chronic kidney disease. Analysis of the modelled AOP network and its topological features determined mitochondrial dysfunction, oxidative stress, and tubular necrosis to be the most connected and central KEs. These KEs can provide a logical foundation for guiding the selection and creation of in vitro assays and in silico tools to substitute for animal-based in vivo experiments in the prediction and assessment of chemical-induced nephrotoxicity in human health.


Subject(s)
Adverse Outcome Pathways , Animal Experimentation , Drug-Related Side Effects and Adverse Reactions , Renal Insufficiency , Animals , Humans , Risk Assessment/methods
3.
Biofabrication ; 14(4)2022 07 05.
Article in English | MEDLINE | ID: mdl-35700695

ABSTRACT

Despite the increasing incidence of kidney-related diseases, we are still far from understanding the underlying mechanisms of these diseases and their progression. This lack of understanding is partly because of a poor replication of the diseasesin vitro,limited to planar culture. Advancing towards three-dimensional models, hereby we propose coaxial printing to obtain microfibers containing a helical hollow microchannel. These recapitulate the architecture of the proximal tubule (PT), an important nephron segment often affected in kidney disorders. A stable gelatin/alginate-based ink was formulated to allow printability while maintaining structural properties. Fine-tuning of the composition, printing temperature and extrusion rate allowed for optimal ink viscosity that led to coiling of the microfiber's inner channel. The printed microfibers exhibited prolonged structural stability (42 days) and cytocompatibility in culture. Healthy conditionally immortalized PT epithelial cells and a knockout cell model for cystinosis (CTNS-/-) were seeded to mimic two genotypes of PT. Upon culturing for 14 days, engineered PT showed homogenous cytoskeleton organization as indicated by staining for filamentous actin, barrier-formation and polarization with apical markerα-tubulin and basolateral marker Na+/K+-ATPase. Cell viability was slightly decreased upon prolonged culturing for 14 days, which was more pronounced inCTNS-/-microfibers. Finally,CTNS-/-cells showed reduced apical transport activity in the microfibers compared to healthy PT epithelial cells when looking at breast cancer resistance protein and multidrug resistance-associated protein 4. Engineered PT incorporated in a custom-designed microfluidic chip allowed to assess leak-tightness of the epithelium, which appeared less tight inCTNS-/-PT compared to healthy PT, in agreement with itsin vivophenotype. While we are still on the verge of patient-oriented medicine, this system holds great promise for further research in establishing advancedin vitrodisease models.


Subject(s)
Kidney Diseases , Neoplasm Proteins , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Humans , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Neoplasm Proteins/metabolism , Printing, Three-Dimensional
4.
Toxicol Lett ; 295: 179-186, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-29959986

ABSTRACT

Iron can catalyze reactive oxygen species (ROS) formation, causing cellular injury. In systemic iron overload, renal tubular epithelial cells are luminally exposed to high iron levels due to glomerular filtration of increased circulating iron. Reports of tubular dysfunction and iron deposition in ß-thalassemia major support an association between increased chronic iron exposure and renal tubular injury. In acute iron exposure, Nuclear factor-erythroid 2-related factor 2 (Nrf2) may protect from iron-induced injury, whereas chronic renal stress may lead to Nrf2 exhaustion. We studied the cytotoxic mechanisms of chronic iron exposure using human conditionally immortalized proximal tubular epithelial cells (ciPTECs). Long-term iron exposure resulted in iron accumulation, cytosolic ROS formation and increased heme oxygenase 1 (HMOX-1) mRNA expression (all p < 0.001). This was accompanied by nuclear translocation of Nrf2 and induction of its target protein NQO1, which both could be blocked by the Nrf2 inhibitor trigonelline. Interestingly, iron and trigonelline incubation reduced ROS production, but did not affect HMOX-1 mRNA levels. Moreover, ferritin protein and CHOP mRNA expression were induced in combined iron and trigonelline incubated cells (p < 0.05). Together, these findings suggest that chronic iron exposure induces oxidative stress and that exhaustion of the antioxidant Nrf2 pathway may lead to renal injury.


Subject(s)
Alkaloids/pharmacology , Ferric Compounds/toxicity , Iron Overload/chemically induced , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/drug effects , NF-E2-Related Factor 2/antagonists & inhibitors , Oxidative Stress/drug effects , Cell Line , Dose-Response Relationship, Drug , Ferritins/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Iron Overload/metabolism , Iron Overload/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
5.
AAPS J ; 20(5): 86, 2018 07 23.
Article in English | MEDLINE | ID: mdl-30039346

ABSTRACT

Drug-induced kidney injury is often observed in the clinics and can lead to long-term organ failure. In this work, we evaluated a novel in vitro system that aims at detecting whether compounds can cause renal proximal tubule damage in man. For this, we implemented organotypic cultures of human conditionally immortalized proximal tubule epithelial cells overexpressing the organic anion transporter 1 (ciPTEC-OAT1) in a three-channel OrganoPlate under microfluidic conditions. Cells were exposed to four known nephrotoxicants (cisplatin, tenofovir, cyclosporine A, and tobramycin). The effect on cell viability and NAG release into the medium was determined. A novel panel of four miRNAs (mir-21, mir-29a, mir-34a, and mir-192) was selected as potential biomarkers of proximal tubule damage. After nephrotoxicant treatment, miRNA levels in culture medium were earlier indicators than cell viability (WST-8 assay) and outperformed NAG for proximal tubule damage. In particular, mir-29a, mir-34a, and mir-192 were highly reproducible between experiments and across compounds, whereas mir-21 showed more variability. Moreover, similar data were obtained in two different laboratories, underlining the reproducibility and technical transferability of the results, a key requirement for the implementation of novel biomarkers. In conclusion, the selected miRNAs behaved like sensitive biomarkers of damage to tubular epithelial cells caused by several nephrotoxicity mechanisms. This biomarker panel, in combination with the 3D cultures of ciPTEC-OAT1 in the OrganoPlate, represents a novel tool for in vitro nephrotoxicity detection. These results pave the way for the application of miRNAs in longitudinal, time-course in vitro toxicity studies.


Subject(s)
Epithelial Cells/drug effects , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/drug effects , MicroRNAs/genetics , Microfluidic Analytical Techniques , Cell Line, Transformed , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genetic Markers , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , MicroRNAs/metabolism , Proof of Concept Study , Reproducibility of Results , Risk Assessment , Time Factors
6.
J Tissue Eng Regen Med ; 12(7): 1670-1678, 2018 07.
Article in English | MEDLINE | ID: mdl-29764003

ABSTRACT

The accumulation of protein-bound toxins in dialyzed patients is strongly associated with their high morbidity and mortality. The bioartificial kidney device (BAK), containing proximal tubule epithelial cells (PTECs) seeded on functionalized synthetic hollow fibre membranes, may be a powerful solution for the active removal of those metabolites. In an earlier study, we developed an upscaled BAK containing conditionally immortalized human PTEC with functional organic cationic transporter 2. Here, we first extended this development to a BAK device having cells with the organic anionic transporter 1, capable of removing anionic uraemic wastes. We confirmed the quality of the conditionally immortalized human PTEC monolayer by confocal microscopy and paracellular inulin-fluorescein isothiocyanate leakage, as well as by the active transport of anionic toxin, indoxyl sulphate. Furthermore, we assessed the immune safety of our system by measuring the production of relevant cytokines by the cells after lipopolysaccharide stimulation. Upon lipopolysaccharide treatment, we observed a polarized secretion of proinflammatory cytokines by the cells: 10-fold higher in the extraluminal space, corresponding to the urine compartment, as compared with the intraluminal space, corresponding to the blood compartment. To the best of our knowledge, our work is the first to show this favourable cell polarization in a BAK upscaled device.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/metabolism , Lipopolysaccharides/pharmacology , Organic Cation Transporter 2/metabolism , Cell Line, Transformed , Humans , Kidney Tubules, Proximal/cytology
7.
Sci Rep ; 7(1): 10623, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28878224

ABSTRACT

Septic acute kidney injury (AKI) associates with poor survival rates and often requires renal replacement therapy. Glucocorticoids may pose renal protective effects in sepsis via stimulation of mitochondrial function. Therefore, we studied the mitochondrial effects of dexamethasone in an experimental inflammatory proximal tubule epithelial cell model. Treatment of human proximal tubule epithelial cells with lipopolysaccharide (LPS) closely resembles pathophysiological processes during endotoxaemia, and led to increased cytokine excretion rates and cellular reactive oxygen species levels, combined with a reduced mitochondrial membrane potential and respiratory capacity. These effects were attenuated by dexamethasone. Dexamethasone specifically increased the expression and activity of mitochondrial complex V (CV), which could not be explained by an increase in mitochondrial mass. Finally, we demonstrated that dexamethasone acidified the intracellular milieu and consequently reversed LPS-induced alkalisation, leading to restoration of the mitochondrial function. This acidification also provides an explanation for the increase in CV expression, which is expected to compensate for the inhibitory effect of the acidified environment on this complex. Besides the mechanistic insights into the beneficial effects of dexamethasone during renal cellular inflammation, our work also supports a key role for mitochondria in this process and, hence, provides novel therapeutic avenues for the treatment of AKI.


Subject(s)
Dexamethasone/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Nephritis, Interstitial/metabolism , Biomarkers , Cell Line , Cells, Cultured , Citrate (si)-Synthase/metabolism , Endotoxemia/etiology , Endotoxemia/metabolism , Endotoxemia/pathology , Epithelial Cells/pathology , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Kidney Tubules, Proximal/pathology , Lipopolysaccharides/adverse effects , Membrane Potential, Mitochondrial/drug effects , Metabolic Networks and Pathways/drug effects , Nephritis, Interstitial/etiology , Nephritis, Interstitial/pathology , Reactive Oxygen Species/metabolism
8.
Sci Rep ; 6: 26715, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27242131

ABSTRACT

The development of a biotechnological platform for the removal of waste products (e.g. uremic toxins), often bound to proteins in plasma, is a prerequisite to improve current treatment modalities for patients suffering from end stage renal disease (ESRD). Here, we present a newly designed bioengineered renal tubule capable of active uremic toxin secretion through the concerted action of essential renal transporters, viz. organic anion transporter-1 (OAT1), breast cancer resistance protein (BCRP) and multidrug resistance protein-4 (MRP4). Three-dimensional cell monolayer formation of human conditionally immortalized proximal tubule epithelial cells (ciPTEC) on biofunctionalized hollow fibers with maintained barrier function was demonstrated. Using a tailor made flow system, the secretory clearance of human serum albumin-bound uremic toxins, indoxyl sulfate and kynurenic acid, as well as albumin reabsorption across the renal tubule was confirmed. These functional bioengineered renal tubules are promising entities in renal replacement therapies and regenerative medicine, as well as in drug development programs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Kidney Tubules, Proximal/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Organic Anion Transport Protein 1/metabolism , Tissue Engineering , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cell Line , Humans , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Kidney Tubules, Proximal/pathology , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , Organic Anion Transport Protein 1/genetics
9.
Sci Rep ; 5: 16702, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26567716

ABSTRACT

The bioartificial kidney (BAK) aims at improving dialysis by developing 'living membranes' for cells-aided removal of uremic metabolites. Here, unique human conditionally immortalized proximal tubule epithelial cell (ciPTEC) monolayers were cultured on biofunctionalized MicroPES (polyethersulfone) hollow fiber membranes (HFM) and functionally tested using microfluidics. Tight monolayer formation was demonstrated by abundant zonula occludens-1 (ZO-1) protein expression along the tight junctions of matured ciPTEC on HFM. A clear barrier function of the monolayer was confirmed by limited diffusion of FITC-inulin. The activity of the organic cation transporter 2 (OCT2) in ciPTEC was evaluated in real-time using a perfusion system by confocal microscopy using 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide (ASP(+)) as a fluorescent substrate. Initial ASP(+) uptake was inhibited by a cationic uremic metabolites mixture and by the histamine H2-receptor antagonist, cimetidine. In conclusion, a 'living membrane' of renal epithelial cells on MicroPES HFM with demonstrated active organic cation transport was successfully established as a first step in BAK engineering.


Subject(s)
Membranes, Artificial , Organic Cation Transport Proteins/metabolism , Cations/metabolism , Cell Adhesion/drug effects , Cell Line , Cimetidine/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Histamine H2 Antagonists/pharmacology , Humans , Immunohistochemistry , Ion Transport/drug effects , Kidney Tubules, Proximal/cytology , Methylamines/chemistry , Methylamines/metabolism , Organic Cation Transporter 2 , Permeability/drug effects , Pyridinium Compounds/chemistry , Pyridinium Compounds/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism
10.
Br J Pharmacol ; 172(20): 4932-45, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26222228

ABSTRACT

BACKGROUND AND PURPOSE: Recently, two phase-II trials demonstrated improved renal function in critically ill patients with sepsis-associated acute kidney injury treated with the enzyme alkaline phosphatase. Here, we elucidated the dual active effect on renal protection of alkaline phosphatase. EXPERIMENTAL APPROACH: The effect of human recombinant alkaline phosphatase (recAP) on LPS-induced renal injury was studied in Sprague-Dawley rats. Renal function was assessed by transcutaneous measurement of FITC-sinistrin elimination in freely moving, awake rats. The mechanism of action of recAP was further investigated in vitro using conditionally immortalized human proximal tubular epithelial cells (ciPTEC). KEY RESULTS: In vivo, LPS administration significantly prolonged FITC-sinistrin half-life and increased fractional urea excretion, which was prevented by recAP co-administration. Moreover, recAP prevented LPS-induced increase in proximal tubule injury marker, kidney injury molecule-1 expression and excretion. In vitro, LPS-induced production of TNF-α, IL-6 and IL-8 was significantly attenuated by recAP. This effect was linked to dephosphorylation, as enzymatically inactive recAP had no effect on LPS-induced cytokine production. RecAP-mediated protection resulted in increased adenosine levels through dephosphorylation of LPS-induced extracellular ADP and ATP. Also, recAP attenuated LPS-induced increased expression of adenosine A2A receptor. However, the A2A receptor antagonist ZM-241385 did not diminish the effects of recAP. CONCLUSIONS AND IMPLICATIONS: These results indicate that the ability of recAP to reduce renal inflammation may account for the beneficial effect observed in septic acute kidney injury patients, and that dephosphorylation of ATP and LPS are responsible for this protective effect.


Subject(s)
Acute Kidney Injury/metabolism , Alkaline Phosphatase/pharmacology , Protective Agents/pharmacology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Alkaline Phosphatase/therapeutic use , Animals , Cells, Cultured , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fluoresceins/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lipopolysaccharides , Male , Oligosaccharides/metabolism , Protective Agents/therapeutic use , Purines/urine , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Triazines/pharmacology , Triazoles/pharmacology
11.
Biotechnol Adv ; 32(7): 1317-1327, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25135479

ABSTRACT

With the world-wide increase of patients with renal failure, the development of functional renal replacement therapies have gained significant interest and novel technologies are rapidly evolving. Currently used renal replacement therapies insufficiently remove accumulating waste products, resulting in the uremic syndrome. A more preferred treatment option is kidney transplantation, but the shortage of donor organs and the increasing number of patients waiting for a transplant warrant the development of novel technologies. The bioartificial kidney (BAK) is such promising biotechnological approach to replace essential renal functions together with the active secretion of waste products. The development of the BAK requires a multidisciplinary approach and evolves at the intersection of regenerative medicine and renal replacement therapy. Here we provide a concise review embracing a compact historical overview of bioartificial kidney development and highlighting the current state-of-the-art, including implementation of living-membranes and the relevance of extracellular matrices. We focus further on the choice of relevant renal epithelial cell lines versus the use of stem cells and co-cultures that need to be implemented in a suitable device. Moreover, the future of the BAK in regenerative nephrology is discussed.


Subject(s)
Bioprosthesis , Biotechnology/methods , Coculture Techniques/methods , Kidneys, Artificial , Tissue Engineering/methods , Animals , Cell Line , Epithelial Cells , Extracellular Matrix , Humans , Mice , Models, Biological , Stem Cells , Swine
12.
Exp Cell Res ; 323(1): 87-99, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24560744

ABSTRACT

Promising renal replacement therapies include the development of a bioartificial kidney using functional human kidney cell models. In this study, human conditionally immortalized proximal tubular epithelial cell (ciPTEC) lines originating from kidney tissue (ciPTEC-T1 and ciPTEC-T2) were compared to ciPTEC previously isolated from urine (ciPTEC-U). Subclones of all ciPTEC isolates formed tight cell layers on Transwell inserts as determined by transepithelial resistance, inulin diffusion, E-cadherin expression and immunocytochemisty. Extracellular matrix genes collagen I and -IV α1 were highly present in both kidney tissue derived matured cell lines (p<0.001) compared to matured ciPTEC-U, whereas matured ciPTEC-U showed a more pronounced fibronectin I and laminin 5 gene expression (p<0.01 and p<0.05, respectively). Expression of the influx carrier Organic Cation Transporter 2 (OCT-2), and the efflux pumps P-glycoprotein (P-gp), Multidrug Resistance Protein 4 (MRP4) and Breast Cancer Resistance Protein (BCRP) were confirmed in the three cell lines using real-time PCR and Western blotting. The activities of OCT-2 and P-gp were sensitive to specific inhibition in all models (p<0.001). The highest activity of MRP4 and BCRP was demonstrated in ciPTEC-U (p<0.05). Finally, active albumin reabsorption was highest in ciPTEC-T2 (p<0.001), while Na(+)-dependent phosphate reabsorption was most abundant in ciPTEC-U (p<0.01). In conclusion, ciPTEC established from human urine or kidney tissue display comparable functional PTEC specific transporters and physiological characteristics, providing ideal human tools for bioartificial kidney development.


Subject(s)
Bioartificial Organs , Kidney Tubules, Proximal/cytology , Kidneys, Artificial , Urine/cytology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/metabolism , Cadherins/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cell Culture Techniques , Cell Line , Collagen Type I/biosynthesis , Collagen Type I/metabolism , Fibronectins/biosynthesis , Humans , Inulin/metabolism , Multidrug Resistance-Associated Proteins/biosynthesis , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Octamer Transcription Factor-2/antagonists & inhibitors , Octamer Transcription Factor-2/biosynthesis , Octamer Transcription Factor-2/metabolism , Tissue Engineering , Transendothelial and Transepithelial Migration/physiology , Kalinin
13.
Nat Rev Nephrol ; 9(7): 385-98, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23670084

ABSTRACT

In the past 8 years, there has been renewed interest in the role of iron in both acute kidney injury (AKI) and chronic kidney disease (CKD). In patients with kidney diseases, renal tubules are exposed to a high concentration of iron owing to increased glomerular filtration of iron and iron-containing proteins, including haemoglobin, transferrin and neutrophil gelatinase-associated lipocalin (NGAL). Levels of intracellular catalytic iron may increase when glomerular and renal tubular cells are injured. Reducing the excessive luminal or intracellular levels of iron in the kidney could be a promising approach to treat AKI and CKD. Understanding the role of iron in kidney injury and as a therapeutic target requires insight into the mechanisms of iron metabolism in the kidney, the role of endogenous proteins involved in iron chelation and transport, including hepcidin, NGAL, the NGAL receptor and divalent metal transporter 1, and iron-induced toxic effects. This Review summarizes emerging knowledge, which suggests that complex mechanisms of iron metabolism exist in the kidney, modulated directly or indirectly by cellular iron content, inflammation, ischaemia and oxidative stress. The potential exists for prevention and treatment of iron-induced kidney injury by customized iron removal or relocation, aided by detailed insight into the underlying pathological mechanisms.


Subject(s)
Acute Kidney Injury/etiology , Iron Overload/complications , Iron/metabolism , Kidney/metabolism , Renal Insufficiency, Chronic/etiology , Acute Kidney Injury/metabolism , Humans , Iron Overload/metabolism , Renal Insufficiency, Chronic/metabolism
14.
Biochim Biophys Acta ; 1832(1): 142-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23017367

ABSTRACT

During chronic kidney disease (CKD), drug metabolism is affected leading to changes in drug disposition. Furthermore, there is a progressive accumulation of uremic retention solutes due to impaired renal clearance. Here, we investigated whether uremic toxins can influence the metabolic functionality of human conditionally immortalized renal proximal tubule epithelial cells (ciPTEC) with the focus on UDP-glucuronosyltransferases (UGTs) and mitochondrial activity. Our results showed that ciPTEC express a wide variety of metabolic enzymes, including UGTs. These enzymes were functionally active as demonstrated by the glucuronidation of 7-hydroxycoumarin (7-OHC; K(m) of 12±2µM and a V(max) of 76±3pmol/min/mg) and p-cresol (K(m) of 33±13µM and a V(max) of 266±25pmol/min/mg). Furthermore, a wide variety of uremic toxins, including indole-3-acetic acid, indoxyl sulfate, phenylacetic acid and kynurenic acid, reduced 7-OHC glucuronidation with more than 30% as compared with controls (p<0.05), whereas UGT1A and UGT2B protein expressions remained unaltered. In addition, our results showed that several uremic toxins inhibited mitochondrial succinate dehydrogenase (i.e. complex II) activity with more than 20% as compared with controls (p<0.05). Moreover, indole-3-acetic acid decreased the reserve capacity of the electron transport system with 18% (p<0.03). In conclusion, this study shows that multiple uremic toxins inhibit UGT activity and mitochondrial activity in ciPTEC, thereby affecting the metabolic capacity of the kidney during CKD. This may have a significant impact on drug and uremic retention solute disposition in CKD patients.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/metabolism , Kidney/metabolism , Mitochondria/metabolism , Uremia/metabolism , Cell Line , Cresols/metabolism , Drug-Related Side Effects and Adverse Reactions/enzymology , Drug-Related Side Effects and Adverse Reactions/genetics , Electron Transport , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Kidney/enzymology , Mitochondria/enzymology , Mitochondria/genetics , Pharmaceutical Preparations/metabolism , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Umbelliferones/metabolism , Uremia/enzymology , Uremia/genetics
16.
Clin Pharmacol Ther ; 89(4): 587-94, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21368750

ABSTRACT

Thiazolidinediones (TZDs) are associated with fluid retention that has been suggested to be resistant to treatment with loop diuretics. This resistance is thought to be caused by upregulation of renal epithelial sodium channels (ENaCs). In this study, we tested whether these mechanisms are of clinical significance. We conducted a well-controlled study in 12 insulin-resistant nondiabetic participants, who received treatment for 9 weeks with either rosiglitazone at a dosage of 4 mg b.i.d. or placebo. The aim of the study was to investigate whether upregulation of ENaCs by rosiglitazone reduces furosemide's natriuretic response and enhances the response to the ENaC inhibitor amiloride. The natriuretic response to furosemide and amiloride and the amount of α-ENaC in urinary exosomes were quantified. Rosiglitazone neither reduced furosemide-induced natriuresis nor changed furosemide's concentration-effect curve. Furthermore, rosiglitazone did not change either amiloride-induced natriuresis nor the amount of urinary α-ENaC. This study challenges previous findings regarding TZD-related ENaC upregulation and suggests that TZD-induced fluid retention should respond normally to loop diuretics.


Subject(s)
Diuretics/pharmacology , Epithelial Sodium Channels/drug effects , Hypoglycemic Agents/pharmacology , Insulin Resistance , Thiazolidinediones/pharmacology , Adult , Amiloride/pharmacokinetics , Amiloride/pharmacology , Cross-Over Studies , Double-Blind Method , Drug Interactions , Epithelial Sodium Channels/metabolism , Exosomes/metabolism , Female , Furosemide/pharmacokinetics , Furosemide/pharmacology , Humans , Kidney/drug effects , Kidney/metabolism , Male , Middle Aged , Natriuresis/drug effects , Rosiglitazone , Up-Regulation/drug effects
17.
Minerva Urol Nefrol ; 61(4): 373-84, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19816390

ABSTRACT

Acute kidney injury (AKI) is a frequent clinical problem with a high mortality rate, generally caused by ischemic insults. Nevertheless, the kidney has a remarkably high capacity to regenerate after ischemic injury. Tubular cells can restore renal function by proliferation and dedifferentiation into a mesenchymal cell-type, but also stem cells residing in bone marrow (BM) have been suggested to contribute. Considerable progress has been made in the development of different techniques to study the role of BM-derived stem cells in renal regeneration after AKI. Trans-differentiation of BM cells to functional tubular epithelium has been demonstrated previously, however, beneficial effects of BM transplantations may have been accelerated by irradiation of mice prior to transplantation and kidney injury. Recent studies support a paracrine or endocrine role of BM-derived cells, in which an improvement of renal function is observed without direct involvement in tubular epithelial engraftment. On the other hand, BM cells have also shown not to improve renal function despite their tubular engraftment. This review gives an overview of the recent progress in studying the role of BM-derived cells as therapeutic strategy in renal tubular repair after acute injury.


Subject(s)
Acute Kidney Injury/surgery , Bone Marrow Transplantation , Animals , Bone Marrow Cells/physiology , Humans
18.
Scand J Rheumatol ; 37(1): 72-4, 2008.
Article in English | MEDLINE | ID: mdl-18189198

ABSTRACT

Sulfasalazine (SSZ) can induce serological and clinical autoimmune reactions but the occurrence of SSZ-related Wegener's granulomatosis (WG) has not been reported before. We describe two patients with rheumatoid factor (RF)-positive rheumatoid arthritis (RA) who developed biopsy-proven WG with serious organ involvement during SSZ therapy. The pathogenetic mechanism that explains the relationship between SSZ and the occurrence of a de novo anti-neutrophil cytoplasmic antibody (ANCA)-related vasculitis or a flare is discussed. We propose that WG can be a rare complication of SSZ therapy and that this, like other autoimmune adverse events of this drug, is mediated by SSZ-induced apoptosis.


Subject(s)
Disease Progression , Granulomatosis with Polyangiitis/drug therapy , Granulomatosis with Polyangiitis/physiopathology , Sulfasalazine/adverse effects , Sulfasalazine/therapeutic use , Adult , Aged , Antirheumatic Agents/adverse effects , Antirheumatic Agents/therapeutic use , C-Reactive Protein/metabolism , Cyclophosphamide/therapeutic use , Glucocorticoids/therapeutic use , Humans , Methylprednisolone/therapeutic use
19.
Kidney Int ; 73(2): 220-5, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17978814

ABSTRACT

The Breast Cancer Resistance Protein (BCRP/ABCG2) is a transporter restricting absorption and enhancing excretion of many compounds including anticancer drugs. This transporter is highly expressed in many tissues; however, in human kidney, only the mRNA was found in contrast to the mouse kidney, where the transporter is abundant. In bcrp/abcg2((-/-)) mice, the expression of two sterol transporter genes, abcg5 and abcg8, was strongly increased in the kidney, perhaps as a compensatory mechanism to upregulate efflux. We found using immunohistochemical analysis clear localization of BCRP/ABCG2 to the proximal tubule brush border membrane of the human kidney comparable to that of other ABC transporters such as P-glycoprotein/ABCB1, MRP2/ABCC2, and MRP4/ABCC4. Hoechst 33342 dye efflux from primary human proximal tubule cells was significantly reduced by the BCRP/ABCG2 inhibitors fumitremorgin C and nelfinavir. Our study shows that in addition to other apical ABC transporters, BCRP/ABCG2 may be important in renal drug excretion.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Kidney Tubules, Proximal/metabolism , Neoplasm Proteins/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/physiology , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Humans , Immunohistochemistry , Kidney Tubules, Proximal/chemistry , Kidney Tubules, Proximal/cytology , Mice , Multidrug Resistance-Associated Protein 2 , Neoplasm Proteins/analysis , Neoplasm Proteins/physiology , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats
20.
Kidney Int ; 72(10): 1233-41, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17851469

ABSTRACT

The multidrug resistance gene 1 product, P-glycoprotein (P-gp), is expressed in several excretory organs, including the apical membrane of proximal tubules. After inducing acute renal failure, P-gp expression is upregulated and this might be a protective function by pumping out toxicants and harmful products of oxidative stress. We characterized renal function of P-gp knockout mice and studied its consequences in renal ischemic damage. Compared with wild-type mice, knockout mice have a lower glomerular filtration rate and renal plasma flow. An augmented urinary excretion of sodium, numerous amino acids, calcium, glucose, and low molecular weight proteins was observed along with an increased diuresis. A higher lithium plasma clearance in the knockout mice suggested proximal tubular dysfunction. Electron microscopy showed mitochondrial abnormalities in proximal tubular cells that could account for decreased adenosine triphosphate levels in the cortex. After inducing ischemia, wild-type mice showed a decrease in creatinine clearance and severe proximal tubular necrosis. In contrast, knockout mice had no signs of tubular damage. Our data indicate that P-gp knockout mice have impaired renal function but are protected against ischemic renal injury.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , Ischemia/prevention & control , Kidney Tubules, Proximal/physiopathology , Acute Kidney Injury/physiopathology , Amino Acids/urine , Animals , Calcium/urine , Diuresis , Fluorescent Antibody Technique , Glomerular Filtration Rate/genetics , Glycosuria , Immunohistochemistry , Ischemia/etiology , Kidney Tubules, Proximal/ultrastructure , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mitochondria/pathology , Mitochondria/ultrastructure , Proteinuria , Renal Circulation/genetics , Sodium/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...