Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Rev Nephrol ; 20(2): 101-119, 2024 02.
Article in English | MEDLINE | ID: mdl-37857763

ABSTRACT

Chronic kidney disease (CKD) is a major public health concern, underscoring a need to identify pathogenic mechanisms and potential therapeutic targets. Reactive oxygen species (ROS) are derivatives of oxygen molecules that are generated during aerobic metabolism and are involved in a variety of cellular functions that are governed by redox conditions. Low levels of ROS are required for diverse processes, including intracellular signal transduction, metabolism, immune and hypoxic responses, and transcriptional regulation. However, excess ROS can be pathological, and contribute to the development and progression of chronic diseases. Despite evidence linking elevated levels of ROS to CKD development and progression, the use of low-molecular-weight antioxidants to remove ROS has not been successful in preventing or slowing disease progression. More recent advances have enabled evaluation of the molecular interactions between specific ROS and their targets in redox signalling pathways. Such studies may pave the way for the development of sophisticated treatments that allow the selective control of specific ROS-mediated signalling pathways.


Subject(s)
Oxidative Stress , Renal Insufficiency, Chronic , Humans , Reactive Oxygen Species/metabolism , Oxidation-Reduction , Signal Transduction
2.
FASEB J ; 37(9): e23129, 2023 09.
Article in English | MEDLINE | ID: mdl-37606578

ABSTRACT

During peritoneal dialysis (PD), the peritoneum is exposed to a bioincompatible dialysate, deteriorating the tissue and limiting the long-term effectiveness of PD. Peritoneal fibrosis is triggered by chronic inflammation induced by a variety of stimuli, including peritonitis. Exposure to PD fluid alters peritoneal macrophages phenotype. Inflammasome activation triggers chronic inflammation. First, it was determined whether inflammasome activation causes peritoneal deterioration. In the in vivo experiments, the increased expression of the inflammasome components, caspase-1 activity, and concomitant overproduction of IL-1ß and IL-18 were observed in a mouse model of peritoneal fibrosis. ASC-positive and F4/80-positive cells colocalized in the subperitoneal mesothelial cell layer. These macrophages expressed high CD44 levels indicating that the CD44-positive macrophages contribute to developing peritoneal deterioration. Furthermore, intravital imaging of the peritoneal microvasculature demonstrated that the circulating CD44-positive leukocytes may contribute to peritoneal fibrosis. Bone marrow transplantation in ASC-deficient mice suppressed inflammasome activation, thereby attenuating peritoneal fibrosis in a high glucose-based PD solution-injected mouse model. Our results suggest inflammasome activation in CD44-positive macrophages may be involved in developing peritoneal fibrosis. The inflammasome-derived pro-inflammatory cytokines might therefore serve as new biomarkers for developing encapsulating peritoneal sclerosis.


Subject(s)
Peritoneal Fibrosis , Peritonitis , Animals , Mice , Peritoneum , Inflammasomes , Disease Models, Animal , Inflammation
4.
Transplantation ; 106(2): 280-288, 2022 02 01.
Article in English | MEDLINE | ID: mdl-33908383

ABSTRACT

BACKGROUND: Neutrophils play a major role in ischemia/reperfusion injury (IRI) in renal transplantation and acute kidney injury. However, it has been difficult to observe changes in neutrophil dynamics over time in living mice kidney. We investigate neutrophil dynamics in IRI in living mice using novel in vivo multiphoton microscope imaging techniques and characterize the renoprotective effects of a selective phosphodiesterase 5 inhibitor, tadalafil. METHODS: Wild-type and endothelial nitric oxide synthase knockout mice, a model of endothelial dysfunction, were used to establish in vivo real-time imaging in living mouse kidneys. Neutrophils were labeled green with Ly-6G monoclonal antibody, and plasma flow was labeled red with BSA. Tadalafil was administered orally 1 h before surgery. Both kidney pedicles were reperfused after 37°C warm ischemia for 45 min. RESULTS: Our novel approach revealed that neutrophils were trapped in glomerulus within a few minutes after reperfusion. They gradually increased over time and infiltrated neutrophils were observed in the tubular lumen and peritubular capillary. The neutrophils were clearly visualized rolling on peritubular capillary plexus at 3 µm/min. The administration of tadalafil significantly reduced neutrophil influx into the glomerulus in both wild-type and endothelial nitric oxide synthase knockout mice. Reduced neutrophil infiltration in tadalafil groups, which was confirmed by flow cytometry, resulted in histopathologically decreased tubular injury. The expression of vascular cell adhesion molecule 1 and kidney injury molecule 1 was partially prevented by tadalafil. CONCLUSIONS: Use of a novel technique contributed to elucidation of neutrophil dynamics after reperfusion. Tadalafil has a potential for inhibiting neutrophil infiltration in renal IRI.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Animals , Ischemia , Kidney , Mice , Neutrophils/metabolism , Reperfusion , Reperfusion Injury/metabolism , Tadalafil/metabolism , Tadalafil/pharmacology
5.
Kidney Int ; 101(3): 551-562, 2022 03.
Article in English | MEDLINE | ID: mdl-34843756

ABSTRACT

Heart failure is frequently accompanied by kidney failure and co-incidence of these organ failures worsens the mortality in patients with heart failure. Recent clinical observations revealed that increased kidney venous pressure, rather than decreased cardiac output, causes the deterioration of kidney function in patients with heart failure. However, the underlying pathophysiology is unknown. Here, we found that decreased blood flow velocity in peritubular capillaries by kidney congestion and upregulation of endothelial nuclear factor-κB (NF-κB) signaling synergistically exacerbate kidney injury. We generated a novel mouse model with unilateral kidney congestion by constriction of the inferior vena cava between kidney veins. Intravital imaging highlighted the notable dilatation of peritubular capillaries and decreased kidney blood flow velocity in the congestive kidney. Damage after ischemia reperfusion injury was exacerbated in the congestive kidney and accumulation of polymorphonuclear leukocytes within peritubular capillaries was noted at the acute phase after injury. Similar results were obtained in vitro, in which polymorphonuclear leukocytes adhesion on activated endothelial cells was decreased in flow velocity-dependent manner but cancelled by inhibition of NF-κB signaling. Pharmacological inhibition of NF-κB for the mice subjected by both kidney congestion and ischemia reperfusion injury ameliorated the accumulation of polymorphonuclear leukocytes and subsequent exacerbation of kidney injury. Thus, our study demonstrates the importance of decreased blood flow velocity accompanying activated NF-κB signaling in aggravation of kidney injury. Hence, inhibition of NF-κB signaling may be a therapeutic candidate for the vicious cycle between heart and kidney failure with increased kidney venous pressure.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Acute Kidney Injury/therapy , Animals , Endothelial Cells , Humans , Kidney , Mice , NF-kappa B , Reperfusion Injury/complications
6.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502177

ABSTRACT

Chronic kidney disease is a common problem in the elderly and is associated with increased mortality. We have reported on the role of nitric oxide, which is generated from endothelial nitric oxide synthase (eNOS), in the progression of aged kidneys. To elucidate the role of endothelial dysfunction and the lack of an eNOS-NO pathway in ageing kidneys, we conducted experiments using eNOS and ASC-deficient mice. C57B/6 J mice (wild type (WT)), eNOS knockout (eNOS KO), and ASC knockout (ASC KO) mice were used in the present study. Then, eNOS/ASC double-knockout (eNOS/ASC DKO) mice were generated by crossing eNOS KO and ASC KO mice. These mice were sacrificed at 17-19 months old. The Masson positive area and the KIM-1 positive area tended to increase in eNOS KO mice, compared with WT mice, but not eNOS/ASC DKO mice. The COX-positive area was significantly reduced in eNOS KO mice, compared with WT and eNOS/ASC DKO mice. To determine whether inflammasomes were activated in infiltrating macrophages, the double staining of IL-18 and F4/80 was performed. IL-18 and F4/80 were found to be co-localised in the tubulointerstitial areas. Inflammasomes play a pivotal role in inflammaging in ageing kidneys. Furthermore, inflammasome activation may accelerate cellular senescence via mitochondrial dysfunction. The importance of endothelial function as a regulatory mechanism suggests that protection of endothelial function may be a potential therapeutic target.


Subject(s)
Aging , Endothelium/physiopathology , Inflammasomes , Kidney/physiopathology , Mitochondria/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Endothelium/enzymology , Endothelium/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/physiology , Nitric Oxide/metabolism , Vascular Diseases/physiopathology
7.
PLoS One ; 16(5): e0251554, 2021.
Article in English | MEDLINE | ID: mdl-33974681

ABSTRACT

Serum zinc (Zn) levels tend to be low in chronic kidney disease (CKD) patients. This cohort study was conducted to investigate the relationship between zinc deficiency and CKD progression. Patients were classified into two groups based on Zn levels < 60 µg/dl (low-Zn group, n = 160) and ≥ 60 µg/dl (high-Zn group, n = 152). The primary outcome was defined as end-stage kidney disease (ESKD) or death and was examined over a 1-year observation period. Overall, the mean Zn level was 59.6 µg/dl and the median eGFR was 20.3 ml/min/1.73 m2. The incidence of the primary outcome was higher in the low-Zn group (p<0.001). Various Cox proportional hazards models adjusted for baseline characteristics showed higher risks of the primary outcome in the low-Zn group than in the high-Zn group. Competing risks analysis showed that low Zn levels were associated with ESKD but not with death. Moreover, in propensity score-matched analysis, the low-Zn group showed a higher risk of the primary outcome [adjusted hazard ratio 1.81 (95% confidence interval 1.02, 3.24)]. Furthermore, an interaction was observed between Zn and serum albumin levels (interaction p = 0.026). The results of this study indicate that zinc deficiency is a risk factor for CKD progression.


Subject(s)
Hypoalbuminemia/blood , Renal Insufficiency, Chronic/blood , Zinc/deficiency , Aged , Aged, 80 and over , Disease Progression , Female , Glomerular Filtration Rate , Humans , Hypoalbuminemia/etiology , Japan/epidemiology , Kaplan-Meier Estimate , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/mortality , Male , Middle Aged , Propensity Score , Renal Insufficiency, Chronic/complications , Retrospective Studies , Risk Factors , Zinc/physiology
8.
JCI Insight ; 5(19)2020 10 02.
Article in English | MEDLINE | ID: mdl-32870819

ABSTRACT

Lupus nephritis (LN) is a major organ complication and cause of morbidity and mortality in patients with systemic lupus erythematosus (SLE). There is an unmet medical need for developing more efficient and specific, mechanism-based therapies, which depends on improved understanding of the underlying LN pathogenesis. Here we present direct visual evidence from high-power intravital imaging of the local kidney tissue microenvironment in mouse models showing that activated memory T cells originated in immune organs and the LN-specific robust accumulation of the glomerular endothelial glycocalyx played central roles in LN development. The glomerular homing of T cells was mediated via the direct binding of their CD44 to the hyaluronic acid (HA) component of the endothelial glycocalyx, and glycocalyx-degrading enzymes efficiently disrupted homing. Short-course treatment with either hyaluronidase or heparinase III provided long-term organ protection as evidenced by vastly improved albuminuria and survival rate. This glycocalyx/HA/memory T cell interaction is present in multiple SLE-affected organs and may be therapeutically targeted for SLE complications, including LN.


Subject(s)
Endothelium, Vascular/immunology , Glycocalyx/metabolism , Hyaluronoglucosaminidase/administration & dosage , Kidney Glomerulus/immunology , Lupus Nephritis/prevention & control , Polysaccharide-Lyases/administration & dosage , T-Lymphocytes/immunology , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Hyaluronic Acid/metabolism , Immunologic Memory/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
9.
Nephrol Dial Transplant ; 35(5): 773-781, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32221606

ABSTRACT

BACKGROUND: Long-term exposure to bioincompatible peritoneal dialysate causes the loss of mesothelial cells and accumulation of matrix proteins, leading to an increase in the thickness of the submesothelial layer, thereby limiting the long-term effectiveness of peritoneal dialysis (PD). However, the detailed molecular mechanisms underlying the process of peritoneal fibrosis have not been clearly elucidated. Wnt/ß-catenin signaling pathway activation has been suggested to play a pivotal role in the development of organ fibrosis. Moreover, Klotho protein can regulate Wnt/ß-catenin signaling. We examined the role of Klotho protein in reducing peritoneal fibrosis by inhibiting Wnt/ß-catenin signaling. METHODS: The ß-catenin-activated transgenic (BAT) driving expression of nuclear ß-galactosidase reporter transgenic (BAT-LacZ) mice, the alpha-Klotho gene under control of human elongation factor 1 alpha promoter [Klotho transgenic (KLTG) and C57BL/6 background] and C57BL/6 mice [wild-type (WT)] were used. The mice received daily intraperitoneal (i.p.) injections of 4.25% glucose with lactate (PD solution) or saline as a control for 4 weeks. Other mice received daily i.p. injections of the same volume of saline (normal control). RESULTS: After exposure to PD, Wnt signal activation was observed on the peritoneal mesothelial cells in WT-PD mice. The peritoneal fibrosis was also accelerated in WT-PD mice. The protein expression of ß-catenin and Wnt-inducible genes were also remarkably increased in WT-PD mice. On the other hand, KLTG-PD mice attenuated activation of Wnt/ß-catenin signaling after exposure to PD and ameliorated the progression of peritoneal fibrosis. CONCLUSIONS: Overexpression of Klotho protein protects the peritoneal membrane through attenuation of the Wnt/ß-catenin signaling pathway. The availability of recombinant Klotho protein would provide a novel potential therapeutic target in peritoneal fibrosis.


Subject(s)
Glucuronidase/physiology , Peritoneal Fibrosis/therapy , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Humans , Klotho Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/pathology , Wnt Proteins/metabolism , beta Catenin/metabolism
10.
Curr Opin Nephrol Hypertens ; 29(1): 112-118, 2020 01.
Article in English | MEDLINE | ID: mdl-31725011

ABSTRACT

PURPOSE OF REVIEW: Emerging data have demonstrated that sodium-glucose cotransporter-2 (SGLT2) inhibitors prevent cardiovascular events, especially heart failure-associated endpoints. Cardiovascular outcome trials have also suggested their renoprotective effects. One large clinical trial investigated renal primary endpoints and demonstrated that SGLT2 inhibitors slowed the progression of diabetic kidney disease (DKD). This review summarizes clinical trial data on renal outcomes and discusses potential underlying mechanisms. RECENT FINDINGS: The EMPA-REG, CANVAS, and DECLARE-TIMI 58 studies revealed that SGLT2 inhibitors reduce the risk of cardiovascular events and concomitantly suggested that these drugs slow the progression of kidney disease in type 2 diabetes. The CREDENCE trial on patients with high-risk type 2 diabetes and chronic kidney disease demonstrated that canagliflozin treatment reduced the relative risk of a composite outcome, including end-stage kidney disease, serum creatinine doubling, and renal/cardiovascular death, by 30% in these patients. Animal experiments revealed that oxidative stress, inflammation, fibrosis, and tubuloglomerular feedback are underlying renoprotective mechanisms behind SGLT2 inhibitors. SUMMARY: Recent clinical trials have established the renoprotective effects of SGLT2 inhibitors. Further investigations on mechanisms of these renoprotective effects will provide deeper insights and understanding of pathogenetic properties of DKD.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Humans , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
11.
FASEB J ; 33(11): 12253-12263, 2019 11.
Article in English | MEDLINE | ID: mdl-31431054

ABSTRACT

Multiple clinical studies have shown that bardoxolone methyl, a potent activator of nuclear factor erythroid 2-related factor 2 (Nrf2), is effective in increasing glomerular filtration rate in patients with chronic kidney disease. However, whether an Nrf2 activator can protect tubules from proteinuria-induced tubular damage via anti-inflammatory and antioxidative stress mechanisms is unknown. Using an Institute of Cancer Research-derived glomerulonephritis (ICGN) mouse model of nephrosis, we examined the effects of dihydro-CDDO-trifluoroethyl amide (dh404), a rodent-tolerable bardoxolone methyl analog, in protecting the tubulointerstitium; dh404 markedly suppressed tubular epithelial cell damage in the renal interstitium of ICGN mice. The tubular epithelial cells of ICGN mice showed a decrease in the size and number of mitochondria, as well as the breakdown of the crista structure, whereas the number and ultrastructure of mitochondria were maintained by the dh404 treatment. To further determine the effect of dh404 on mitochondrial function, we used human proximal tubular cells in vitro. Stimulation with albumin and free fatty acid increased mitochondrial reactive oxygen species (ROS). However, dh404 administration diminished mitochondrial ROS. Our data show that dh404 significantly reduced proteinuria-induced tubular cell mitochondrial damage, suggesting that improved redox balance and mitochondrial function and suppression of inflammation underlie the cytoprotective mechanism of Nrf2 activators, including bardoxolone methyl, in diabetic kidney disease.-Nagasu, H., Sogawa, Y., Kidokoro, K., Itano, S., Yamamoto, T., Satoh, M., Sasaki, T., Suzuki, T., Yamamoto, M., Wigley, W. C., Proksch, J. W., Meyer, C. J., Kashihara, N. Bardoxolone methyl analog attenuates proteinuria-induced tubular damage by modulating mitochondrial function.


Subject(s)
Kidney Tubules, Proximal/drug effects , Mitochondria/drug effects , Oleanolic Acid/analogs & derivatives , Proteinuria/complications , Animals , Cells, Cultured , Humans , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred ICR , Mitochondria/physiology , NF-E2-Related Factor 2/physiology , Oleanolic Acid/pharmacology , Reactive Oxygen Species/metabolism
12.
Circulation ; 140(4): 303-315, 2019 07 23.
Article in English | MEDLINE | ID: mdl-30773020

ABSTRACT

BACKGROUND: Sodium glucose cotransporter 2 inhibitors may reduce kidney hyperfiltration, thereby preventing diabetic kidney disease progression, which may in turn reduce cardiovascular risk, including heart failure. However, the mechanisms that regulate renal function responses to sodium glucose cotransporter 2 inhibition are not yet fully understood. We explored the renal protective effects of sodium glucose cotransporter 2 inhibition with empagliflozin, with a focus on glomerular hemodynamic effects and tubuloglomerular feedback using in vivo multiphoton microscopy imaging techniques. METHODS: C57BL/6 mice and spontaneously diabetic Ins2+/Akita mice were studied. The mice were treated with empagliflozin (20 mg·kg-1·d-1) and insulin for 4 weeks, and the single-nephron glomerular filtration rate was measured using multiphoton microscope. A neuronal nitric oxide synthase inhibitor (7-nitroindazole, 20 mg·kg-1·d-1) or a cyclooxygenase-2 inhibitor (SC58236, 6 mg/L), or an A1 adenosine receptor antagonist (8-cyclopentyl-1,3-dipropylxanthine, 1 mg·kg-1·d-1) was administered to elucidate the mechanisms of tubuloglomerular feedback signaling and single-nephron glomerular filtration rate regulation. RESULTS: The urinary excretion of adenosine, nitric oxide metabolites, and the prostanoid prostaglandin E2 was also quantified. The single-nephron glomerular filtration rate in the Ins2+/Akita group was higher than in controls (C57BL/6; 4.9±1.3 nL/min versus Ins2+/Akita; 15.8±6.8 nL/min) and lower in Ins2+/Akita /empagliflozin to 8.0±3.3 nL/min (P<0.01). In vivo imaging also revealed concomitant afferent arteriolar dilation (P<0.01) and increased glomerular permeability of albumin in the Ins2+/Akita group. Empagliflozin ameliorated these changes (P<0.01). Urinary adenosine excretion in the Ins2+/Akita/empagliflozin group was higher than in Ins2+/Akita (Ins2+/Akita; 3.4±1.4 nmol/d, Ins2+/Akita/empagliflozin; 11.2±3.0 nmol/d, P<0.05), whereas nitric oxide metabolites and prostaglandin E2 did not differ. A1 adenosine receptor antagonism, but not neuronal nitric oxide synthase or cyclooxygenase-2 inhibition, blocked the effect of empagliflozin on renal function. Empagliflozin increased urinary adenosine excretion and reduced hyperfiltration via afferent arteriolar constriction, effects that were abolished by A1 adenosine receptor blockade. CONCLUSIONS: Adenosine/A1 adenosine receptor pathways play a pivotal role in the regulation of the single-nephron glomerular filtration rate via tubuloglomerular feedback mechanisms in response to sodium glucose cotransporter 2 inhibition, which may contribute to renal and cardiovascular protective effects reported in clinical trials.


Subject(s)
Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/drug therapy , In Vitro Techniques/methods , Kidney/physiopathology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Animals , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/therapeutic use , Glucosides/pharmacology , Glucosides/therapeutic use , Humans , Male , Mice , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
13.
Nephrology (Carlton) ; 24(1): 28-38, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29068550

ABSTRACT

AIM: Acute kidney injury (AKI) is associated with chronic kidney disease, as well as high mortality, but effective treatments for AKI are still lacking. A recent study reported the prevention of renal injury, such as ischemia-reperfusion injury, by 5-aminolevulinic acid (ALA), which induces an antioxidant effect. The current study aimed to investigate the effect of ALA in a rhabdomyolysis-induced mouse model of AKI created by intramuscular injection of 50% glycerol. METHODS: Rhabdomyolysis-induced AKI was induced by an intramuscular injection of glycerol (5 mL/kg body weight) into mice. Administration of ALA (30 mg/kg, by gavage) was started from 48 h before or 24 h after glycerol injection. The mice were sacrificed at 72 h after glycerol injection. The roles of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), which is one of the Nrf2-related antioxidants, were further investigated through in vivo and in vitro methods. RESULTS: 5-aminolevulinic acid markedly reduced renal dysfunction and tubular damage in mice with rhabdomyolysis-induced AKI. ALA administration decreased oxidative stress, macrophage infiltration, and inflammatory cytokines and apoptosis. The expression of Nrf2 was upregulated by ALA administration. However, administration of Zinc protoporphyrin-9 (ZnPPIX) to inhibit HO-1 activity did not abolish these improvements by ALA. The expression of Nrf2-associated antioxidant factors other than HO-1 was also increased. CONCLUSION: These findings indicate that ALA exerts its antioxidant activity via Nrf2-associated antioxidant factors to provide a renoprotective effect against rhabdomyolysis-induced AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Aminolevulinic Acid/pharmacology , Antioxidants/pharmacology , Kidney Tubules/drug effects , NF-E2-Related Factor 2/agonists , Rhabdomyolysis/prevention & control , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Cells, Cultured , Cytokines/metabolism , Cytoprotection , Disease Models, Animal , Glycerol , Heme Oxygenase-1/metabolism , Humans , Inflammation Mediators/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Rhabdomyolysis/chemically induced , Rhabdomyolysis/metabolism , Rhabdomyolysis/pathology , Signal Transduction/drug effects
14.
PLoS One ; 13(10): e0203823, 2018.
Article in English | MEDLINE | ID: mdl-30281670

ABSTRACT

Hypertension causes vascular complications, such as stroke, cardiovascular disease, and chronic kidney disease (CKD). The relationship between endothelial dysfunction and progression of kidney disease is well known. However, the relationship between the eNOS-NO pathway and chronic inflammation, which is a common pathway for the progression of kidney disease, remains unexplored. We performed in vivo experiments to determine the role of the eNOS-NO pathway by using eNOS-deficient mice in a hypertensive kidney disease model. All mice were unilateral nephrectomized (Nx). One week after Nx, the mice were randomly divided into two groups: the aldosterone infusion groups and the vehicle groups. All mice also received a 1% NaCl solution instead of drinking water. The aldosterone infusion groups were treated with hydralazine to correct blood pressure differences. After four weeks of drug administration, all mice were euthanized, and blood and kidney tissue samples were collected. In the results, NLRP3 inflammasome activation was elevated in the kidneys of the eNOS-deficient mice, and tubulointerstitial fibrosis was accelerated. Suppression of inflammasome activation by knocking out ASC prevented tubulointerstitial injury in the eNOS knockout mice, indicating that the eNOS-NO pathway is involved in the development of kidney dysfunction through acceleration of NLRP3 inflammasome in macrophages. We revealed that endothelial function, particularly the eNOS-NO pathway, attenuates the progression of renal tubulointerstitial injury via suppression of inflammasome activation. Clinically, patients who develop vascular endothelial dysfunction have lifestyle diseases, such as hypertension or diabetes, and are known to be at risk for CKD. Our study suggests that the eNOS-NO pathway could be a therapeutic target for the treatment of chronic kidney disease associated with endothelial dysfunction.


Subject(s)
Hypertension, Renal/metabolism , Hypertension/metabolism , Inflammasomes/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Renal Insufficiency, Chronic/metabolism , Aldosterone/pharmacology , Animals , Antihypertensive Agents/administration & dosage , Disease Models, Animal , Endothelium/pathology , Endothelium/physiopathology , Fibrosis , Humans , Hydralazine/administration & dosage , Hypertension/complications , Hypertension, Renal/drug therapy , Hypertension, Renal/pathology , Inflammasomes/drug effects , Kidney/pathology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nitric Oxide Synthase Type III/genetics , Primary Cell Culture , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology , Vasodilator Agents/administration & dosage
15.
Sci Rep ; 7(1): 8801, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821730

ABSTRACT

Chronic inflammation can be a major driver of the failure of a variety of organs, including chronic kidney disease (CKD). The NLR family pyrin domain-containing 3 (NLRP3) inflammasome has been shown to play a pivotal role in inflammation in a mouse kidney disease model. Nuclear factor erythroid 2-related factor 2 (Nrf2), the master transcription factor for anti-oxidant responses, has also been implicated in inflammasome activation under physiological conditions. However, the mechanism underlying inflammasome activation in CKD remains elusive. Here, we show that the loss of Nrf2 suppresses fibrosis and inflammation in a unilateral ureter obstruction (UUO) model of CKD in mice. We consistently observed decreased expression of inflammation-related genes NLRP3 and IL-1ß in Nrf2-deficient kidneys after UUO. Increased infiltration of M1, but not M2, macrophages appears to mediate the suppression of UUO-induced CKD symptoms. Furthermore, we found that activation of the NLRP3 inflammasome is attenuated in Nrf2-deficient bone marrow-derived macrophages. These results demonstrate that Nrf2-related inflammasome activation can promote CKD symptoms via infiltration of M1 macrophages. Thus, we have identified the Nrf2 pathway as a promising therapeutic target for CKD.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , NF-E2-Related Factor 2/deficiency , Ureteral Obstruction/etiology , Ureteral Obstruction/metabolism , Animals , Disease Models, Animal , Fibrosis , Gene Expression Profiling , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Kidney/metabolism , Kidney/pathology , Macrophage Activation/genetics , Macrophage Activation/immunology , Mice , Mice, Knockout , Ureteral Obstruction/pathology , Ureteral Obstruction/therapy
16.
J Renin Angiotensin Aldosterone Syst ; 17(2): 1470320315625704, 2016.
Article in English | MEDLINE | ID: mdl-27216079

ABSTRACT

INTRODUCTION: We investigated the feasibility of using a fluorescence resonance energy transfer system to image enzymatic activity in order to evaluate the effects of aliskiren (a direct renin inhibitor) on diabetic nephropathy. MATERIALS AND METHODS: First, we induced diabetes in C57BL/6J mice using streptozotocin, then treated them with either aliskiren (25 mg/kg/day) or the angiotensin type 1 receptor blocker valsartan (15 mg/kg/day) for four weeks. Finally, we utilized renin fluorescence resonance energy transfer substrate to assess renin activity. RESULTS: Renin activity was much higher in the kidneys of diabetic mice compared to those of the non-diabetic control mice. While aliskiren inhibited this activity, valsartan did not. We noted that production of reactive oxygen species intensified and the bioavailability of nitric oxide diminished in the glomeruli of diabetic mice. Aliskiren and valsartan significantly ameliorated these effects. They suppressed glomerular production of reactive oxygen species and urinary albumin excretion. In fact, urinary albumin excretion in diabetic mice treated with aliskiren or valsartan was lower than that in untreated diabetic mice. Furthermore, aliskiren and valsartan significantly reduced glomerular permeability by maintaining the glomerular endothelial surface layer. CONCLUSION: Fluorescence resonance energy transfer could provide a new tool for evaluating tissue and plasma enzymatic activity.


Subject(s)
Amides/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Fluorescence Resonance Energy Transfer , Fumarates/pharmacology , Imaging, Three-Dimensional , Kidney/drug effects , Albuminuria/blood , Albuminuria/complications , Albuminuria/drug therapy , Amides/therapeutic use , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Feasibility Studies , Fumarates/therapeutic use , Kidney Glomerulus/drug effects , Macromolecular Substances/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Permeability/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Renin/blood , Superoxides/metabolism
17.
Curr Opin Nephrol Hypertens ; 25(3): 168-73, 2016 May.
Article in English | MEDLINE | ID: mdl-27008595

ABSTRACT

PURPOSE OF REVIEW: The review aims to provide a brief summary and evaluation of the current state of research that uses multiphoton fluorescence microscopy for intravital kidney imaging. RECENT FINDINGS: Direct visualization of the glomerular filter, proximal and distal tubule segments, and the renal vasculature in the living, intact kidney in zebrafish, mouse, and rat models with high temporal and spatial resolution provided new insights into the function of the normal and diseased kidney. New technical developments in fluorescence excitation and detection, in combination with transgenic animal models for cell function and fate mapping, and serial imaging of the same glomerulus in the same animal over several days further advanced the field of nephrology research, and the understanding of disease mechanisms. SUMMARY: Intravital multiphoton imaging has solved many critical technical barriers in kidney research and allowed the dynamic portrayal of the structure and function of various renal cell types in vivo. It has become a widely used research technique, with significant past achievements, and tremendous potential for future development and applications for the study and better understanding of kidney diseases.


Subject(s)
Intravital Microscopy , Kidney Diseases/diagnosis , Kidney/pathology , Microscopy, Fluorescence, Multiphoton , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Kidney/blood supply , Kidney Diseases/pathology , Microscopy, Fluorescence, Multiphoton/methods , Models, Animal
18.
Lab Invest ; 96(1): 25-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26552047

ABSTRACT

Increased generation of reactive oxygen species (ROS) is a common denominative pathogenic mechanism underlying vascular and renal complications in diabetes mellitus. Endothelial NAD(P)H oxidase is a major source of vascular ROS, and it has an important role in endothelial dysfunction. We hypothesized that activation of endothelial NAD(P)H oxidase initiates and worsens the progression of diabetic nephropathy, particularly in the development of albuminuria. We used transgenic mice with endothelial-targeted overexpression of the catalytic subunit of NAD(P)H oxidase, Nox2 (NOX2TG). NOX2TG mice were crossed with Akita insulin-dependent diabetic (Akita) mice that develop progressive hyperglycemia. We compared the progression of diabetic nephropathy in Akita versus NOX2TG-Akita mice. NOX2TG-Akita mice and Akita mice developed significant albuminuria above the baseline at 6 and 10 weeks of age, respectively. Compared with Akita mice, NOX2TG-Akita mice exhibited higher levels of NAD(P)H oxidase activity in glomeruli, developed glomerular endothelial perturbations, and attenuated expression of glomerular glycocalyx. Moreover, in contrast to Akita mice, the NOX2TG-Akita mice had numerous endothelial microparticles (blebs), as detected by scanning electron microscopy, and increased glomerular permeability. Furthermore, NOX2TG-Akita mice exhibited distinct phenotypic changes in glomerular mesangial cells expressing α-smooth muscle actin, and in podocytes expressing increased levels of desmin, whereas the glomeruli generated increased levels of ROS. In conclusion, activation of endothelial NAD(P)H oxidase in the presence of hyperglycemia initiated and exacerbated diabetic nephropathy characterized by the development of albuminuria. Moreover, ROS generated in the endothelium compounded glomerular dysfunctions by altering the phenotypes of mesangial cells and compromising the integrity of the podocytes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , NADPH Oxidases/metabolism , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/enzymology , Diabetic Nephropathies/enzymology , Female , Glomerular Filtration Rate/physiology , Kidney Glomerulus/chemistry , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Mice , Podocytes/chemistry , Reactive Oxygen Species/metabolism
19.
Kidney Int ; 88(1): 44-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25738253

ABSTRACT

Intravital imaging using multiphoton microscopy (MPM) has become an increasingly popular and widely used experimental technique in kidney research over the past few years. MPM allows deep optical sectioning of the intact, living kidney tissue with submicron resolution, which is unparalleled among intravital imaging approaches. MPM has solved a long-standing critical technical barrier in renal research to study several complex and inaccessible cell types and anatomical structures in vivo in their native environment. Comprehensive and quantitative kidney structure and function MPM studies helped our better understanding of the cellular and molecular mechanisms of the healthy and diseased kidney. This review summarizes recent in vivo MPM studies with a focus on the glomerulus and the filtration barrier, although select, glomerulus-related renal vascular and tubular functions are also mentioned. The latest applications of serial MPM of the same glomerulus in vivo, in the intact kidney over several days, during the progression of glomerular disease are discussed. This visual approach, in combination with genetically encoded fluorescent markers of cell lineage, has helped track the fate and function (e.g., cell calcium changes) of single podocytes during the development of glomerular pathologies, and provided visual proof for the highly dynamic, rather than static, nature of the glomerular environment. Future intravital imaging applications have the promise to further push the limits of optical microscopy, and to advance our understanding of the mechanisms of kidney injury. Also, MPM will help to study new mechanisms of tissue repair and regeneration, a cutting-edge area of kidney research.


Subject(s)
Glomerular Filtration Barrier/anatomy & histology , Glomerular Filtration Barrier/physiology , Intravital Microscopy/methods , Kidney Diseases/pathology , Microscopy, Fluorescence, Multiphoton , Animals , Calcium/metabolism , Cell Movement , Kidney Diseases/physiopathology , Mice , Podocytes/physiology , Rats , Time-Lapse Imaging , Zebrafish
20.
J Am Soc Nephrol ; 24(7): 1139-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23620395

ABSTRACT

In diabetes, endothelial nitric oxide synthase (eNOS) produces superoxide anion rather than nitric oxide, referred to as "eNOS uncoupling," which may contribute to endothelial dysfunction, albuminuria, and diabetic nephropathy. Reduced levels of endothelium-derived tetrahydrobiopterin (BH4), an essential cofactor for eNOS, promote eNOS uncoupling. Accelerated degradation of guanosine triphosphate cyclohydrolase I (GTPCH I), the rate-limiting enzyme in BH4 biosynthesis, also occurs in diabetes, suggesting that GTPCH I may have a role in diabetic microvascular disease. Here, we crossed endothelium-dominant GTPCH I transgenic mice with Ins2(+/Akita) diabetic mice and found that endothelial overexpression of GTPCH I led to higher levels of intrarenal BH4 and lower levels of urinary albumin and reactive oxygen species compared with diabetic control mice. Furthermore, GTPCH I overexpression attenuated the hyperpermeability of macromolecules observed in diabetic control mice. In addition, we treated Ins2(+/Akita) mice with metformin, which activates AMP-activated protein kinase (AMPK) and thereby slows the degradation of GTPCH I; despite blood glucose levels that were similar to untreated mice, those treated with metformin had significantly less albuminuria. Similarly, in vitro, treating human glomerular endothelial cells with AMPK activators attenuated glucose-induced reductions in phospho-AMPK, GTPCH I, and coupled eNOS. Taken together, these data suggest that maintenance of endothelial GTPCH I expression and the resulting improvement in BH4 biosynthesis ameliorate diabetic nephropathy.


Subject(s)
Biopterins/analogs & derivatives , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , GTP Cyclohydrolase/metabolism , Albuminuria/metabolism , Animals , Biopterins/metabolism , Cell Line , Endothelium, Vascular/metabolism , Humans , Metformin/pharmacology , Mice , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...