Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Clin Med ; 9(9)2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32971813

ABSTRACT

(1) Background: Arterial hypertension (HTN) is one of the most relevant cardiovascular risk factors. Nowadays multiple pharmaceutical treatment options exist with novel interventional methods (e.g., baroreflex activation therapy (BAT)) as a last resort to treat patients with resistant HTN. Although pathophysiology behind resistant HTN is still not fully understood. There is evidence that selected biomarkers may be involved in the pathophysiology of HTN. (2) Methods: We investigated serum SDC4-levels in patients suffering from resistant HTN before and 6 months after BAT implantation. We collected 19 blood samples from patients with resistant HTN and blood pressure above target and measured serum SDC4-levels. (3) Results: Our results showed high serum SDC4-levels in patients with resistant HTN as compared to a healthy population. Patients with both, resistant HTN and diabetes mellitus type II, demonstrated higher serum SDC4-levels. ß-blockers had lowering effects on serum SDC4-levels, whereas calcium channel blockers were associated with higher levels of serum SDC4. BAT implantation did not lead to a significant difference in serum SDC4-levels after 6 months of therapy. (4) Conclusion: Based on our results we propose SDC4 is elevated in patients suffering from resistant HTN. Thus, SDC4 might be a potential marker for endothelial dysfunction in patients with resistant hypertension.

2.
Nephrol Dial Transplant ; 34(1): 49-62, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29726981

ABSTRACT

Background: Our laboratory has previously demonstrated that Sirt1endo-/- mice show endothelial dysfunction and exaggerated renal fibrosis, whereas mice with silenced endothelial transforming growth factor beta (TGF-ß) signaling are resistant to fibrogenic signals. Considering the fact that the only difference between these mutant mice is confined to the vascular endothelium, this indicates that secreted substances contribute to these contrasting responses. Methods: We performed an unbiased proteomic analysis of the secretome of renal microvascular endothelial cells (RMVECs) isolated from these two mutants. We cultured renal fibroblasts and RMVECs and used microfluidic devices for coculturing. Results: Dickkopf-3 (DKK3), a putative ligand of the Wnt/ß-catenin pathway, was present exclusively in the fibrogenic secretome. In cultured fibroblasts, DKK3 potently induced myofibroblast activation. In addition, DKK3 antagonized effects of DKK1, a known inhibitor of the Wnt pathway, in conversion of fibroblasts to myofibroblasts. In RMVECs, DKK3 induced endothelial-mesenchymal transition and impaired their angiogenic competence. The inhibition of endothelial outgrowth, enhanced myofibroblast formation and endothelial-mesenchymal transition were confirmed in coculture. In reporter DKK3-eGFP × Col3.6-GFPcyan mice, DKK3 was marginally expressed under basal conditions. Adriamycin-induced nephropathy resulted in upregulation of DKK3 expression in tubular and, to a lesser degree, endothelial compartments. Sulindac sulfide was found to exhibit superior Wnt pathway-suppressive action and decreased DKK3 signals and the extent of renal fibrosis. Conclusions: In conclusion, this unbiased proteomic screen of the profibrogenic endothelial secretome revealed DKK3 acting as an agonist of the Wnt pathway, enhancing formation of myofibroblasts and endothelial-mesenchymal transition and impairing angiogenesis. A potent inhibitor of the Wnt pathway, sulindac sulfide, suppressed nephropathy-induced DKK3 expression and renal fibrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Endothelium, Vascular/pathology , Epithelial-Mesenchymal Transition , Fibrosis/pathology , Kidney Diseases/pathology , Proteome/analysis , Receptor, Transforming Growth Factor-beta Type II/physiology , Sirtuin 1/physiology , Animals , Endothelium, Vascular/metabolism , Fibrosis/metabolism , Kidney Diseases/metabolism , Mice , Mice, Knockout , Proteomics , Wnt Signaling Pathway , beta Catenin/metabolism
3.
Dev Biol ; 443(1): 78-91, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30189195

ABSTRACT

Maternal undernutrition during pregnancy (MUN) often leads to low birth weight (LBW) neonates that have a reduced total nephron endowment, leaving these neonates susceptible to kidney disease throughout their lives. For reasons unknown, these LBW neonates have impaired kidney development due to a severe reduction in renal SIX2+ stem cells during nephrogenesis. Using a mouse model of MUN, we investigated SIX2+ stem cell reduction in the LBW neonate. Significant upregulation of the protein fetuin-B (measured by PCR and immunoblotting) in the MUN mother's placenta, organs and circulation yielded a 3-fold increase of this protein in the embryonic kidney. Recombinant fetuin-B, administered to healthy pregnant mothers at the concentration equivalent to that in the MUN mother, crossed the placenta and reduced both SIX2+ stem cells by 50% and nephron formation by 66% in embryonic kidneys (measured by immunofluorescence and the physical dissector/fractionator stereological method). Administration of fetuin-B to kidney explants yielded similar reductions in renal SIX2+ stem cells and nephron formation. Fetuin-B treatment of isolated embryonic renal SIX2+ stem cell primary cultures 1) increased NF-kB activity and apoptosis, 2) reduced cell proliferation due to upregulated p21 nuclear activity and subsequent cell cycle arrest, and 3) enhanced generation of reactive oxygen species (measured by fluorescence microscopy). In conclusion, MUN increases fetuin-B in the developing embryonic kidney. The increase in fetuin-B blunts nephrogenesis by reducing SIX2+ stem cells by promoting their apoptosis (via NF-kB upregulation), blunting their proliferative renewal (via p21 upregulation) and enhancing oxidative stress.


Subject(s)
Fetal Nutrition Disorders/metabolism , Fetuin-B/metabolism , Kidney/embryology , Animals , Apoptosis/physiology , Embryonic Stem Cells/metabolism , Female , Fetal Nutrition Disorders/genetics , Homeodomain Proteins/metabolism , Infant, Low Birth Weight/physiology , Kidney/metabolism , Male , Maternal Health , Mice , Nephrons/embryology , Nephrons/metabolism , Oxidative Stress/physiology , Pregnancy , Primary Cell Culture , Transcription Factors/metabolism , Up-Regulation
4.
Am J Physiol Renal Physiol ; 315(3): F583-F594, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29846114

ABSTRACT

Maternal undernutrition (MUN) during pregnancy leads to low-birth weight (LBW) neonates that have a reduced kidney nephron endowment and higher morbidity as adults. Using a severe combined caloric and protein-restricted mouse model of MUN to generate LBW mice, we examined the progression of renal insufficiency in LBW adults. Through 6 mo of age, LBW males experienced greater albuminuria (ELISA analysis), a more rapid onset of glomerular hypertrophy, and a worse survival rate than LBW females. In contrast, both sexes experienced a comparable progressive decline in renal vascular density (immunofluorescence analysis), renal blood flow (Laser-Doppler flowmetry analysis), glomerular filtration rate (FITC-sinistrin clearance analysis), and a progressive increase in systemic blood pressure (measured via tail-cuff method). Isolated aortas from both LBW sexes demonstrated reduced vasodilation in response to ACh, indicative of reduced nitric oxide bioavailability and endothelial dysfunction. ELISA and immunofluorescence analysis revealed a significant increase of circulating reactive oxygen species and NADPH oxidase type 4 (NOX4) expression in both LBW sexes, although these increases were more pronounced in males. Although more effective in males, chronic tempol treatment did improve all observed pathologies in both sexes of LBW mice. Chronic NOX4 inhibition with GKT137831 was more effective than tempol in preventing pathologies in LBW males. In conclusion, despite some minor differences, LBW female and male adults have a reduced nephron endowment comparable with progressive renal and vascular dysfunction, which is associated with increased oxidative stress and subsequent endothelial dysfunction. Tempol treatment and/or NOX4 inhibition attenuates renal and vascular dysfunction in LBW adults.


Subject(s)
Birth Weight , Glomerular Filtration Rate , Kidney Diseases/physiopathology , Kidney/physiopathology , Malnutrition/physiopathology , Oxidative Stress , Prenatal Exposure Delayed Effects , Age Factors , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Antioxidants/pharmacology , Caloric Restriction , Cyclic N-Oxides/pharmacology , Diet, Protein-Restricted , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Glomerular Filtration Rate/drug effects , Hemodynamics , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Male , Malnutrition/metabolism , Maternal Nutritional Physiological Phenomena , Mice , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/metabolism , Oxidative Stress/drug effects , Pregnancy , Pyrazoles/pharmacology , Pyrazolones , Pyridines/pharmacology , Pyridones , Renal Circulation , Spin Labels
5.
Am J Physiol Heart Circ Physiol ; 314(3): H484-H496, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29101181

ABSTRACT

Syndecan-4 (Synd4) is a member of the membrane-spanning, glycocalyx-forming proteoglycan family. It has been suggested that Synd4 participates in renal fibrosis. We compared wild-type and fibrosis-prone endothelial sirtuin 1-deficient (Sirt1endo-/-) mice, the latter being a model of global endothelial dysfunction. We performed mass spectrometry analysis, which revealed that Synd4 was highly enriched in the secretome of renal microvascular endothelial cells obtained from Sirt1endo-/- mice upon stimulation with transforming growth factor-ß1; notably, all detectable peptides were confined to the ectodomain of Synd4. Elevated Synd4 was due to enhanced NF-κB signaling in Sirt1endo-/- mice, while its shedding occurred as a result of oxidative stress in Sirt1 deficiency. Synd4 expression was significantly enhanced after unilateral ureteral obstruction compared with contralateral kidneys. Furthermore, hyperplasia of renal myofibroblasts accompanied by microvascular rarefaction and overexpression of Synd4 were detected in Sirt1endo-/- mice. The ectodomain of Synd4 acted as a chemoattractant for monocytes with higher levels of macrophages and higher expression levels of Synd4 in the extracellular matrix of Sirt1endo-/- mice. In vitro, ectodomain application resulted in generation of myofibroblasts from cultured renal fibroblasts, while in vivo, subcapsular injection of ectodomain increased interstitial fibrosis. Moreover, the endothelial glycocalyx was reduced in Sirt1endo-/- mice, highlighting the induction of Synd4 occurring in parallel with the depletion of its intact form and accumulation of its ectodomain in Sirt1endo-/- mice. On the basis of our experimental results, we propose that it is the Synd4 ectodomain per se that is partially responsible for fibrosis in unilateral ureteral obstruction, especially when it is combined with endothelial dysfunction. NEW & NOTEWORTHY Our findings suggest that endothelial dysfunction induces the expression of syndecan-4 via activation of the NF-κB pathway. Furthermore, we show that syndecan-4 is shed to a greater amount because of increased oxidative stress in dysfunctional endothelial cells and that the release of the syndecan-4 ectodomain leads to tubulointerstitial fibrosis.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Kidney Diseases/metabolism , Kidney/blood supply , Microvessels/metabolism , Syndecan-4/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Fibrosis , Glycocalyx/metabolism , Hyperplasia , Kidney/metabolism , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Mice, Knockout , Microvessels/pathology , Microvessels/physiopathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , NF-kappa B/metabolism , Oxidative Stress , Protein Domains , Signal Transduction , Sirtuin 1/deficiency , Sirtuin 1/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
6.
Redox Biol ; 13: 600-607, 2017 10.
Article in English | MEDLINE | ID: mdl-28806702

ABSTRACT

During sepsis, the alarmin HMGB1 is released from tissues and promotes systemic inflammation that results in multi-organ damage, with the kidney particularly susceptible to injury. The severity of inflammation and pro-damage signaling mediated by HMGB1 appears to be dependent on the alarmin's redox state. Therefore, we examined HMGB1 redox in kidney cells during sepsis. Using intravital microscopy, CellROX labeling of kidneys in live mice indicated increased ROS generation in the kidney perivascular endothelium and tubules during lipopolysaccharide (LPS)-induced sepsis. Subsequent CellROX and MitoSOX labeling of LPS-stressed endothelial and kidney proximal tubule cells demonstrated increased ROS generation in these cells as sepsis worsens. Consequently, HMGB1 oxidation increased in the cytoplasm of kidney cells during its translocation from the nucleus to the circulation, with the degree of oxidation dependent on the severity of sepsis, as measured in in vivo mouse samples using a thiol assay and mass spectrometry (LC-MS/MS). The greater the oxidation of HMGB1, the greater the ability of the alarmin to stimulate pro-inflammatory cyto-/chemokine release (measured by Luminex Multiplex) and alter mitochondrial ATP generation (Luminescent ATP Detection Assay). Administration of glutathione and thioredoxin inhibitors to cell cultures enhanced HMGB1 oxidation during sepsis in endothelial and proximal tubule cells, respectively. In conclusion, as sepsis worsens, ROS generation and HMGB1 oxidation increases in kidney cells, which enhances HMGB1's pro-inflammatory signaling. Conversely, the glutathione and thioredoxin systems work to maintain the protein in its reduced state.


Subject(s)
HMGB1 Protein/metabolism , Oxidative Stress , Sepsis/metabolism , Adenosine Triphosphate/metabolism , Animals , Cytokines/metabolism , HMGB1 Protein/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kidney Tubules, Proximal/cytology , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Signal Transduction
7.
Pediatr Res ; 82(2): 340-348, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28419086

ABSTRACT

BackgroundLow birth weight (LBW) neonates have impaired kidney development that leaves them susceptible to kidney disease and hypertension during adulthood. The study here identifies events that blunt nephrogenesis and kidney development in the murine LBW neonate.MethodsWe examined survival, kidney development, GFR, gene expression, and cyto-/chemokines in the LBW offspring of malnourished (caloric and protein-restricted) pregnant mice.ResultsMalnourished pregnant mothers gave birth to LBW neonates that had 40% reduced body weight and 54% decreased survival. Renal blood perfusion was reduced by 37%, whereas kidney volume and GFR were diminished in the LBW neonate. During gestation, the LBW neonatal kidney had 2.2-fold increased apoptosis, 76% decreased SIX2+ progenitor cells, downregulation of mesenchymal-to-epithelial signaling factors Wnt9b and Fgf8, 64% less renal vesicle formation, and 32% fewer nephrons than controls. At birth, increased plasma levels of IL-1ß, IL-6, IL-12(p70), and granulocyte-macrophage colony-stimulating factor in the LBW neonate reduced SIX2+ progenitor cells.ConclusionIncreased pro-inflammatory cytokines in the LBW neonate decrease SIX2+ stem cells in the developing kidney. Reduced renal stem cells (along with the decreased mesenchymal-to-epithelial signaling) blunt renal vesicle generation, nephron formation, and kidney development. Subsequently, the mouse LBW neonate has reduced glomeruli volume, renal perfusion, and GFR.


Subject(s)
Animals, Newborn , Infant, Low Birth Weight , Kidney/growth & development , Animals , Chemokines/blood , Cytokines/blood , Female , Gene Expression , Glomerular Filtration Rate , Kidney/metabolism , Kidney/physiology , Mice , Pregnancy
8.
Stem Cells Transl Med ; 6(3): 992-1005, 2017 03.
Article in English | MEDLINE | ID: mdl-28297566

ABSTRACT

Accumulation of myofibroblasts is a hallmark of renal fibrosis. A significant proportion of myofibroblasts has been reported to originate via endothelial-mesenchymal transition. We initially hypothesized that exposing myofibroblasts to the extract of endothelial progenitor cells (EPCs) could reverse this transition. Indeed, in vitro treatment of transforming growth factor-ß1 (TGF-ß1)-activated fibroblasts with EPC extract prevented expression of α-smooth muscle actin (α-SMA); however, it did not enhance expression of endothelial markers. In two distinct models of renal fibrosis-unilateral ureteral obstruction and chronic phase of folic acid-induced nephropathy-subcapsular injection of EPC extract to the kidney prevented and reversed accumulation of α-SMA-positive myofibroblasts and reduced fibrosis. Screening the composition of EPC extract for cytokines revealed that it is enriched in leukemia inhibitory factor (LIF) and vascular endothelial growth factor. Only LIF was capable of reducing fibroblast-to-myofibroblast transition of TGF-ß1-activated fibroblasts. In vivo subcapsular administration of LIF reduced the number of myofibroblasts and improved the density of peritubular capillaries; however, it did not reduce the degree of fibrosis. A receptor-independent ligand for the gp130/STAT3 pathway, hyper-interleukin-6 (hyper-IL-6), not only induced a robust downstream increase in pluripotency factors Nanog and c-Myc but also exhibited a powerful antifibrotic effect. In conclusion, EPC extract prevented and reversed fibroblast-to-myofibroblast transition and renal fibrosis. The component of EPC extract, LIF, was capable of preventing development of the contractile phenotype of activated fibroblasts but did not eliminate TGF-ß1-induced collagen synthesis in cultured fibroblasts and models of renal fibrosis, whereas a receptor-independent gp130/STAT3 agonist, hyper-IL-6, prevented fibrosis. In summary, these studies, through the evolution from EPC extract to LIF and then to hyper-IL-6, demonstrate the instructive role of microenvironmental cues and may provide in the future a facile strategy to prevent and reverse renal fibrosis. Stem Cells Translational Medicine 2017;6:992-1005.


Subject(s)
Cellular Microenvironment , Kidney/pathology , 3T3 Cells , Animals , Cellular Microenvironment/drug effects , Chemokines/metabolism , Cytokine Receptor gp130/metabolism , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibrosis , Interleukin-6/pharmacology , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myofibroblasts/cytology , Myofibroblasts/drug effects , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Ureteral Obstruction/pathology
9.
Antioxid Redox Signal ; 25(3): 119-46, 2016 07 20.
Article in English | MEDLINE | ID: mdl-26906267

ABSTRACT

SIGNIFICANCE: A common link between all forms of acute and chronic kidney injuries, regardless of species, is enhanced generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) during injury/disease progression. While low levels of ROS and RNS are required for prosurvival signaling, cell proliferation and growth, and vasoreactivity regulation, an imbalance of ROS and RNS generation and elimination leads to inflammation, cell death, tissue damage, and disease/injury progression. RECENT ADVANCES: Many aspects of renal oxidative stress still require investigation, including clarification of the mechanisms which prompt ROS/RNS generation and subsequent renal damage. However, we currently have a basic understanding of the major features of oxidative stress pathology and its link to kidney injury/disease, which this review summarizes. CRITICAL ISSUES: The review summarizes the critical sources of oxidative stress in the kidney during injury/disease, including generation of ROS and RNS from mitochondria, NADPH oxidase, and inducible nitric oxide synthase. The review next summarizes the renal antioxidant systems that protect against oxidative stress, including superoxide dismutase and catalase, the glutathione and thioredoxin systems, and others. Next, we describe how oxidative stress affects kidney function and promotes damage in every nephron segment, including the renal vessels, glomeruli, and tubules. FUTURE DIRECTIONS: Despite the limited success associated with the application of antioxidants for treatment of kidney injury/disease thus far, preventing the generation and accumulation of ROS and RNS provides an ideal target for potential therapeutic treatments. The review discusses the shortcomings of antioxidant treatments previously used and the potential promise of new ones. Antioxid. Redox Signal. 25, 119-146.


Subject(s)
Disease Susceptibility , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney/metabolism , Oxidants/metabolism , Oxidative Stress , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Humans , Kidney/pathology , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Tubules/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Mitochondria/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Renal Circulation
10.
Am J Pathol ; 186(2): 248-58, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26683662

ABSTRACT

Sepsis is a systemic inflammatory syndrome induced by bacterial infection that can lead to multiorgan failure. Endothelial surface glycocalyx (ESG) decorating the inner wall of blood vessels is a regulator of multiple vascular functions. Here, we tested a hypothesis that patchy degradation of ESG occurs early in sepsis and is a result of exocytosis of lysosome-related organelles. Time-lapse video microscopy revealed that exocytosis of Weibel-Palade bodies and secretory lysosomes occurred a few minutes after application of lipopolysaccharides to endothelial cells. Two therapeutic maneuvers, a nitric oxide intermediate, NG-hydroxy-l-arginine, and culture media conditioned by endothelial progenitor cells reduced the motility of lysosome-related organelles. Confocal and stochastic optical reconstruction microscopy confirmed the patchy loss of ESG simultaneously with the exocytosis of lysosome-related organelles and Weibel-Palade bodies in cultured endothelial cells and mouse aorta. The loss of ESG was blunted by pretreatment with NG-hydroxy-l-arginine or culture media conditioned by endothelial progenitor cells. Moreover, these treatments resulted in a significant reduction in deaths of septic mice. Our data support the hypothesis assigning to stress-induced exocytosis of these organelles the role of a hair-trigger for local degradation of ESG that initiates leukocyte infiltration, increase in vascular permeability, and partially accounts for the later rates of morbidity and mortality.


Subject(s)
Exocytosis/drug effects , Glycocalyx/metabolism , Sepsis/metabolism , Animals , Capillary Permeability/drug effects , Cell Line , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hair/drug effects , Hair/metabolism , Humans , Lipopolysaccharides/pharmacology , Lysosomes/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Sepsis/drug therapy
11.
Stem Cells Transl Med ; 4(7): 852-61, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25947337

ABSTRACT

UNLABELLED: : We previously reported the delivery of endothelial progenitor cells (EPCs) embedded in hyaluronic acid-based (HA)-hydrogels protects renal function during acute kidney injury (AKI) and promotes angiogenesis. We attempted to further ameliorate renal dysfunction by coembedding EPCs with renal mesenchymal stem cells (MSCs), while examining their paracrine influence on cytokine/chemokine release and proinflammatory macrophages. A live/dead assay determined whether EPC-MSC coculturing improved viability during lipopolysaccharide (LPS) treatment, and HA-hydrogel-embedded delivery of cells to LPS-induced AKI mice was assessed for effects on mean arterial pressure (MAP), renal blood flow (RBF), circulating cytokines/chemokines, serum creatinine, proteinuria, and angiogenesis (femoral ligation). Cytokine/chemokine release from embedded stem cells was examined, including effects on macrophage polarization and release of proinflammatory molecules. EPC-MSC coculturing improved stem cell viability during LPS exposure, an effect augmented by MSC hypoxic preconditioning. The delivery of coembedded EPCs with hypoxic preconditioned MSCs to AKI mice demonstrated additive improvement (compared with EPC delivery alone) in medullary RBF and proteinuria, with comparable effects on serum creatinine, MAP, and angiogenesis. Exposure of proinflammatory M1 macrophages to EPC-MSC conditioned medium changed their polarization to anti-inflammatory M2. Incubation of coembedded EPCs-MSCs with macrophages altered their release of cytokines/chemokines, including enhanced release of anti-inflammatory interleukin (IL)-4 and IL-10. EPC-MSC delivery to endotoxemic mice elevated the levels of circulating M2 macrophages and reduced the circulating cytokines/chemokines. In conclusion, coembedding EPCs-MSCs improved their resistance to stress, impelled macrophage polarization from M1 to M2 while altering their cytokine/chemokines release, reduced circulating cytokines/chemokines, and improved renal and vascular function when MSCs were hypoxically preconditioned. SIGNIFICANCE: This report provides insight into a new therapeutic approach for treatment of sepsis and provides a new and improved strategy using hydrogels for the delivery of stem cells to treat sepsis and, potentially, other injuries and/or diseases. The delivery of two different stem cell lines (endothelial progenitor cells and mesenchymal stem cells; delivered alone and together) embedded in a protective bioengineered scaffolding (hydrogel) offers many therapeutic benefits for the treatment of sepsis. This study shows how hydrogel-delivered stem cells elicit their effects and how hydrogel embedding enhances the therapeutic efficacy of delivered stem cells. Hydrogel-delivered stem cells influence the components of the overactive immune system during sepsis and work to counterbalance the release of many proinflammatory and prodamage substances from immune cells, thereby improving the associated vascular and kidney damage.

12.
Prostaglandins Other Lipid Mediat ; 120: 17-20, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25936481

ABSTRACT

We are witnessing the emergence of a novel type of biological regulation, namely, the communication between cells via their secreted substances, the secretome. This brief overview is based on the available published data and our own experience. We discuss three vignettes illustrating the importance of communication via the secretome: (1) the secretome of stem cells and its effects in sepsis and systemic inflammatory response; (2) the profibrotic secretomes partially responsible for development of fibrotic complications; and (3) the contribution of senescence-associated secretory products to the propagation of the senescence phenotype. Considering the richness of secretomes of different cells under diverse conditions, it becomes imperative to gain insights into their individual components in an attempt to harness cell secretomes for therapeutic purposes.


Subject(s)
Cell Communication , Cells/cytology , Cells/metabolism , Animals , Cells/pathology , Humans , Ischemia/pathology , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Sepsis/pathology
13.
Physiol Rep ; 3(4)2015 Apr.
Article in English | MEDLINE | ID: mdl-25896979

ABSTRACT

We sought to characterize a minor renal cryoinjury that allows investigation into renal damage processes and subsequent endogenous repair mechanisms. To achieve this, we induced a small cryoinjury to mice, in which the transient superficial application of a liquid nitrogen-cooled cryoprobe to the exposed kidney induces a localized lesion that did not impair renal function. The resulting cryoinjury was examined by immunohistochemistry and Laser-Doppler flowmetry. Within hours of cryoinjury induction, tubular and vascular necrotic damage was observed, while blood flow in the directly injured area was reduced by 65%. The injured area demonstrated a peak in tubular and perivascular cell proliferation at 4 days postinjury, while apoptosis and fibrosis peaked at day 7. Infiltration of macrophages into the injury was first observed at day 4, and peaked at day 7. Vascular density in the direct injured area was lowest at day 7. As compared to the direct injured area, the (peripheral) penumbral region surrounding the directly injured area demonstrated enhanced cellular proliferation (2.5-6-fold greater), vascular density (1.6-2.9 fold greater) and blood perfusion (twofold greater). After 4 weeks, the area of damage was reduced by 73%, fibrosis decreased by 50% and blood flow in the direct injured area was reestablished by 63% with almost complete perfusion restoration in the injury's penumbral region. In conclusion, kidney cryoinjury provides a flexible facile model for the study of renal damage and associated endogenous repair processes.

14.
Kidney Int ; 87(1): 95-108, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24940804

ABSTRACT

High-mobility group box 1 (HMGB1) undergoes acetylation, nuclear-to-cytoplasmic translocation, and release from stressed kidneys, unleashing a signaling cascade of events leading to systemic inflammation. Here, we tested whether the deacetylase activity of Sirtuin1 (SIRT1) participates in regulating nuclear retention of HMGB1 to ultimately modulate damage signaling initiated by HMGB1 secretion during stress. When immunoprecipitated acetylated HMGB1 was incubated with SIRT1, HMGB1 acetylation decreased by 57%. Proteomic analysis showed that SIRT1 deacetylates HMGB1 at four lysine residues (55, 88, 90, and 177) within the proinflammatory and nuclear localization signal domains of HMGB1. Genetic ablation or pharmacological inhibition of SIRT1 in endothelial cells increased HMGB1 acetylation and translocation. In vivo, deletion of SIRT1 reduced nuclear HMGB1 while increasing its acetylation and release into circulation during basal and ischemic conditions, causing increased renal damage. Conversely, resveratrol pretreatment led to decreased HMGB1 acetylation, its nuclear retention, decreased systemic release, and reduced tubular damage. Thus, a vicious cycle is set into motion in which the inflammation-induced repression of SIRT1 disables deacetylation of HMGB1, facilitates its nuclear-to-cytoplasmic translocation, and systemic release, thereby maintaining inflammation.


Subject(s)
HMGB1 Protein/metabolism , Sirtuin 1/physiology , Acetylation , Animals , Cells, Cultured , Endothelial Cells , Humans , Mice
15.
PLoS One ; 9(2): e87853, 2014.
Article in English | MEDLINE | ID: mdl-24504266

ABSTRACT

We recently demonstrated the use of in vitro expanded kidney-derived mesenchymal stem cells (KMSC) protected peritubular capillary endothelial cells in acute renal ischemia-reperfusion injury. Herein, we isolated and characterized microparticles (MPs) from KMSC. We investigated their in vitro biologic effects on human endothelial cells and in vivo renoprotective effects in acute ischemia-reperfusion renal injury. MPs were isolated from the supernatants of KMSC cultured in anoxic conditions in serum-deprived media for 24 hours. KMSC-derived MPs demonstrated the presence of several adhesion molecules normally expressed on KMSC membranes, such as CD29, CD44, CD73, α4, 5, and 6 integrins. Quantitative real time PCR confirmed the presence of 3 splicing variants of VEGF-A (120, 164, 188), bFGF and IGF-1 in isolated MPs. MPs labeled with PKH26 red fluorescence dye were incorporated by cultured human umbilical vein endothelial cells (HUVEC) via surface molecules such as CD44, CD29, and α4, 5, and 6 integrins. MP dose dependently improved in vitro HUVEC proliferation and promoted endothelial tube formation on growth factor reduced Matrigel. Moreover, apoptosis of human microvascular endothelial cell was inhibited by MPs. Administration of KMSC-derived MPs into mice with acute renal ischemia was followed by selective engraftment in ischemic kidneys and significant improvement in renal function. This was achieved by improving proliferation, of peritubular capillary endothelial cell and amelioration of peritubular microvascular rarefaction. Our results support the hypothesis that KMSC-derived MPs may act as a source of proangiogenic signals and confer renoprotective effects in ischemic kidneys.


Subject(s)
Acute Kidney Injury/metabolism , Cell-Derived Microparticles/metabolism , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Apoptosis , Biological Transport , Cell Proliferation , Cell-Derived Microparticles/ultrastructure , Disease Models, Animal , Gene Expression Profiling , Gene Transfer, Horizontal , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Neovascularization, Physiologic , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
16.
Hypertension ; 63(3): e46-52, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24379188

ABSTRACT

NFAT5 is a transcription factor that protects the kidney from hypertonic stress and also is activated by hypoxia. We hypothesized that NFAT5 mitigates the extent of renal damage induced by ischemia-reperfusion injury (IRI). Mice were subjected to IRI by unilateral clamping of the left renal pedicle for 30 minutes followed by reperfusion. After 3 hours of reperfusion, the level of NFAT5 mRNA was similar in contralateral and clamped kidneys. However, after 48 hours, NFAT5 mRNA accumulation increased ≈3-fold in both outer medulla and medullary thick ascending limb tubules. NFAT1 levels were elevated at 3 hours but did not increase further at 48 hours. Mice were then either pretreated for 72 hours with an intrarenal injection of a lentivirus short-hairpin RNA construct to silence NFAT5 (enhanced green fluorescent protein-U6-N5-ex8) or a control vector (enhanced green fluorescent protein-U6) before induction of IRI. Neutrophil gelatinase-associated lipocalin and kidney ischemia molecule-1 mRNA levels increased after IRI and further increased after knockdown of NFAT5, suggesting that silencing of NFAT5 exacerbates renal damage during IRI. In contrast, silencing of NFAT1 had no effect on the levels of neutrophil gelatinase-associated lipocalin or kidney ischemia molecule-1 mRNA. Hematoxylin and eosin staining revealed patchy denudation of renal epithelial cells and tubular dilation when NFAT5 was silenced. The number of TUNEL-positive cells in the outer and inner medulla of the clamped kidney increased nearly 2-fold after knockdown of NFAT5 and was associated with an increase in the number of caspase-3-positive cells. Collectively, the data suggest that NFAT5 is part of a protective mechanism that limits renal damage induced by IRI.


Subject(s)
Acute Kidney Injury/metabolism , Gene Expression Regulation , RNA, Messenger/genetics , Reperfusion Injury/complications , Transcription Factors/genetics , Acute Kidney Injury/etiology , Animals , Apoptosis , Blotting, Western , Disease Models, Animal , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reperfusion Injury/metabolism , Transcription Factors/metabolism
17.
J Am Soc Nephrol ; 25(2): 276-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24136919

ABSTRACT

Sirtuin 1 (SIRT1) depletion in vascular endothelial cells mediates endothelial dysfunction and premature senescence in diverse cardiovascular and renal diseases. However, the molecular mechanisms underlying these pathologic effects remain unclear. Here, we examined the phenotype of a mouse model of vascular senescence created by genetically ablating exon 4 of Sirt1 in endothelial cells (Sirt1(endo-/-)). Under basal conditions, Sirt1(endo-/-) mice showed impaired endothelium-dependent vasorelaxation and angiogenesis, and fibrosis occurred spontaneously at low levels at an early age. In contrast, induction of nephrotoxic stress (acute and chronic folic acid-induced nephropathy) in Sirt1(endo-/-) mice resulted in robust acute renal functional deterioration followed by an exaggerated fibrotic response compared with control animals. Additional studies identified matrix metalloproteinase-14 (MMP-14) as a target of SIRT1. In the kidneys of Sirt1(endo-/-) mice, impaired angiogenesis, reduced matrilytic activity, and retention of the profibrotic cleavage substrates tissue transglutaminase and endoglin accompanied MMP-14 suppression. Furthermore, restoration of MMP-14 expression in SIRT1-depeleted mice improved angiogenic and matrilytic functions of the endothelium, prevented renal dysfunction, and attenuated nephrosclerosis. Our findings establish a novel mechanistic molecular link between endothelial SIRT1 depletion, downregulation of MMP-14, and the development of nephrosclerosis.


Subject(s)
Matrix Metalloproteinase 14/physiology , Nephrosclerosis/enzymology , Sirtuin 1/deficiency , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Animals , Cellular Senescence , Concanavalin A/pharmacology , Down-Regulation , Endothelium, Vascular/physiopathology , Exons/genetics , Extracellular Matrix/metabolism , Fibrosis , Folic Acid/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Human Umbilical Vein Endothelial Cells , Kidney/enzymology , Kidney/pathology , Kidney/physiopathology , Kidney Failure, Chronic/chemically induced , Kidney Failure, Chronic/metabolism , Male , Matrix Metalloproteinase 14/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , Neovascularization, Physiologic , Nephrosclerosis/genetics , Nephrosclerosis/pathology , Regeneration , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Sirtuin 1/physiology , Vasodilation
18.
PLoS One ; 8(6): e65458, 2013.
Article in English | MEDLINE | ID: mdl-23776484

ABSTRACT

Endothelial Cell Dysfunction (ECD) is a recognized harbinger of a host of chronic cardiovascular diseases. Using a mouse model of ECD triggered by treatment with L-Nω-methylarginine (L-NMMA), we previously demonstrated that renal microvasculature displays a perturbed protein profile, including diminished expression of two key enzymes of the Krebs cycle associated with a Warburg-type suppression of mitochondrial metabolism. We hypothesized that supplementation with L-glutamine (GLN), that can enter the Krebs cycle downstream this enzymatic bottleneck, would normalize vascular function and alleviate mitochondrial dysfunction. To test this hypothesis, mice with chronic L-NMMA-induced ECD were co-treated with GLN at different concentrations for 2 months. Results confirmed that L-NMMA led to a defect in acetylcholine-induced relaxation of aortic rings that was dose-dependently prevented by GLN. In caveolin-1 transgenic mice characterized by eNOS inactivation, L-NMMA further impaired vasorelaxation which was partially rescued by GLN co-treatment. Pro-inflammatory profile induced by L-NMMA was blunted in mice co-treated with GLN. Using an LC/MS platform for metabolite profiling, we sought to identify metabolic perturbations associated with ECD and offset by GLN supplementation. 3453 plasma molecules could be detected with 100% frequency in mice from at least one treatment group. Among these, 37 were found to be differentially expressed in a 4-way comparison of control vs. LNMMA both with and without GLN. One of such molecules, hippuric acid, an "uremic toxin" was found to be elevated in our non-uremic mice receiving L-NMMA, but normalized by treatment with GLN. Ex vivo analysis of hippuric acid effects on vasomotion demonstrated that it significantly reduced acetylcholine-induced vasorelaxation of vascular rings. In conclusion, functional and metabolic profiling of animals with early ECD revealed macrovasculopathy and that supplementation GLN is capable of improving vascular function. Metabolomic analyses reveal elevation of hippuric acid, which may further exacerbate vasculopathy even before the development of uremia.


Subject(s)
Acetylcholine/pharmacology , Aorta/drug effects , Endothelial Cells/drug effects , Glutamine/pharmacology , Vasodilation/drug effects , omega-N-Methylarginine/pharmacology , Animals , In Vitro Techniques , Male , Mice
19.
Biomatter ; 3(1)2013.
Article in English | MEDLINE | ID: mdl-23507925

ABSTRACT

Adoptive transfer of stem cells has shown potential as an effective treatment for acute kidney injury (AKI). The current strategy for adoptive transfer of stem cells is by intravenous injection. However, this conventional method of stem cell delivery is riddled with problems causing reduced efficacy of the therapeutic potential of delivered stem cells. This review summarizes the recent advancements in an alternative method of stem cell delivery for treatment of AKI, embedding stem cells in hyaluronic acid (HA-) based hydrogels followed by their implantation. Furthermore, one stem cell type in particular, endothelial progenitor cells (EPC), have shown remarkable therapeutic benefits for treatment of AKI when delivered by HA-hydrogels. The review also summarizes the delivery of EPC by HA-hydrogels in the setting of AKI.


Subject(s)
Acute Kidney Injury/therapy , Endothelial Progenitor Cells/transplantation , Hyaluronic Acid/administration & dosage , Hydrogels/chemistry , Acute Kidney Injury/physiopathology , Animals , Cell- and Tissue-Based Therapy/methods , Humans , Hydrogels/administration & dosage , Tissue Embedding , Tissue Scaffolds
20.
J Am Soc Nephrol ; 24(4): 529-36, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23349311

ABSTRACT

The list of signals sent by an injured organ to systemic circulation, so-called danger signals, is growing to include multiple metabolites and secreted moieties, thus revealing a highly complex and integrated network of interlinked systemic proinflammatory and proregenerative messages. Emerging new data indicate that, apart from the well established local inflammatory response to AKI, danger signaling unleashes a cascade of precisely timed, interdependent, and intensity-gradated mediators responsible for development of the systemic inflammatory response. This fledgling realization of the importance of the systemic inflammatory response to the localized injury and inflammation is at the core of this brief overview. It has a potential to explain the additive effects of concomitant diseases or preexisting chronic conditions that can prime the systemic inflammatory response and exacerbate it out of proportion to the actual degree of acute kidney damage. Although therapies for ameliorating AKI per se remain limited, a potentially powerful strategy that could reap significant benefits in the future is to modulate the intensity of danger signals and consequently the systemic inflammatory response, while preserving its intrinsic proregenerative stimuli.


Subject(s)
Acute Kidney Injury/pathology , Kidney/pathology , Systemic Inflammatory Response Syndrome/pathology , Acute Kidney Injury/metabolism , Humans , Kidney/metabolism , Signal Transduction , Systemic Inflammatory Response Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...