Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Am J Transl Res ; 13(9): 10014-10037, 2021.
Article in English | MEDLINE | ID: mdl-34650679

ABSTRACT

To date, the treatment of acute kidney injury (AKI) remains a difficult problem for clinicians. In the present study, we assessed whether ZLN005, a novel peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) agonist, can protect against ischemic AKI in vivo and in vitro. Notably, ZLN005 treatment significantly alleviated Ischemia-reperfusion (I/R)-induced tubular injury and reversed the decrease in hypoxia-reoxygenation-induced cell viability by restoring PGC-1α expression in a dose-dependent manner. This beneficial effect of ZLN005 was associated with the preservation of mitochondrial fatty acid oxidation (MitoFAO) and the alleviation of oxidative stress. Cotreatment with etomoxir, a specific inhibitor of carnitine palmitoyltransferase-1α (CPT-1α) activity, or CPT-1α siRNA abrogated ZLN005-induced antistress responses by mitigating reactive oxygen species production and decreasing apoptosis under ischemia-hypoxia conditions by suppressing MitoFAO. Further studies revealed that activation of endoplasmic reticulum (ER) stress may be involved in the effect of CPT-1α inhibition observed in vivo and in vitro. Collectively, our results suggest that ZLN005 confers a protective effect on I/R-induced kidney injury by mitigating ER stress through the restoration of MitoFAO by targeting PGC-1α.

2.
Redox Biol ; 36: 101671, 2020 09.
Article in English | MEDLINE | ID: mdl-32829253

ABSTRACT

In the present study, we hypothesized that hypoxia-inducible factor 1α (HIF-1α)-mediated mitophagy plays a protective role in ischemia/reperfusion (I/R)-induced acute kidney injury (AKI). Mitophagy was evaluated by measuring the changes of mitophagy flux, mitochondria DNA copy number, and the changes of mitophagy-related proteins including translocase of outer mitochondrial membrane 20 (TOMM20), cytochrome c oxidase IV (COX IV), microtubule-associated protein 1 light chain 3B (LC3B), and mitochondria adaptor nucleoporin p62 in HK2 cells, a human tubular cell line. Results show that HIF-1α knockout significantly attenuated hypoxia/reoxygenation (H/R)-induced mitophagy, aggravated H/R-induced apoptosis, and increased the production of reactive oxygen species (ROS). Similarly, H/R induced significantly increase in Bcl-2 19-kDa interacting protein 3 (BNIP3), a downstream regulator of HIF-1α. Notably, BNIP3 overexpression reversed the inhibitory effect of HIF-1α knockout on H/R-induced mitophagy, and prevented the enhancing effect of HIF-1α knockout on H/R-induced apoptosis and ROS production. For in vivo study, we established HIF-1αflox/flox; cadherin-16-cre mice in which tubular HIF-1α was specifically knockout. It was found that tubular HIF-1α knockout significantly inhibited I/R-induced mitophagy, and aggravated I/R-induced tubular apoptosis and kidney damage. In contrast, adenovirus-mediated BNIP3 overexpression significantly reversed the decreased mitophagy, and prevented enhanced kidney damage in tubular HIF-1α knockout mice with I/R injury. In summary, our study demonstrated that HIF-1α-BNIP3-mediated mitophagy in tubular cells plays a protective role through inhibition of apoptosis and ROS production in acute kidney damage.


Subject(s)
Mitophagy , Reperfusion Injury , Animals , Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/metabolism , Kidney/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism
3.
J Clin Hypertens (Greenwich) ; 22(5): 914-923, 2020 05.
Article in English | MEDLINE | ID: mdl-32271999

ABSTRACT

Hypertensive crises are associated with high rates of target organ complications and poor outcomes. A recent shift from the definition of malignant hypertension to hypertension-multiorgan damage (MOD) contributes to the diagnosis and management of hypertensive crises. Here, we prospectively included 166 adult (≥18 years old) patients with hypertensive crises (blood pressure >180/120 mm Hg). Target organs and causes of hypertension were assessed. Patients who were diagnosed with malignant hypertensive retinopathy, the absence of malignant hypertensive retinopathy but the presence of damage to at least 3 organs, and the absence of both retinopathy and MOD were classified as the malignant hypertension (n = 48), hypertension-MOD (n = 42), and hypertension without MOD (n = 76) groups, respectively. Patients were followed to evaluate renal and cardiovascular prognoses. At baseline, patients with malignant hypertension had worse renal function, higher level of albuminuria, and more severe microvascular damage than those with hypertension-MOD. Both had similar proportions of malignant arteriolar nephrosclerosis (83% vs 64%), left ventricular hypertrophy (90% vs 88%), abnormal repolarization (71% vs 60%), and left ventricular dysfunction (12% vs 21%). At the twenty months of follow-up, both the malignant hypertension and hypertension-MOD groups had similar blood pressure control rates and proteinuria. Both groups had worse renal outcomes than the hypertension without MOD group (P = .002). Patients with hypertension-MOD (HR = 0.67, [95% CI: 0.30-1.46], P = .31) had similar renal event-free survival than patients with MHT after adjustments of age, sex, blood pressure, and proteinuria control. These results suggest that in hypertensive crises, both malignant hypertension and hypertension-MOD have impact on adverse renal outcomes.


Subject(s)
Hypertension , Adolescent , Adult , Humans , Follow-Up Studies , Hypertension/complications , Hypertension/epidemiology , Prospective Studies
4.
J Cereb Blood Flow Metab ; 40(12): 2374-2386, 2020 12.
Article in English | MEDLINE | ID: mdl-31865842

ABSTRACT

Endothelial progenitor cell transplantation is a potential therapeutic approach in brain ischemia. However, whether the therapeutic effect of endothelial progenitor cells is via affecting complement activation is unknown. We established a mouse focal ischemia model (n = 111) and transplanted endothelial progenitor cells into the peri-infarct region immediately after brain ischemia. Neurological outcomes and brain infarct/atrophy volume were examined after ischemia. Expression of C3, C3aR and pro-inflammatory factors were further examined to explore the role of endothelial progenitor cells in ischemic brain. We found that endothelial progenitor cells improved neurological outcomes and reduced brain infarct/atrophy volume after 1 to 14 days of ischemia compared to the control (p < 0.05). C3 and C3aR expression in the brain was up-regulated at 1 day up to 14 days (p < 0.05). Endothelial progenitor cells reduced astrocyte-derived C3 (p < 0.05) and C3aR expression (p < 0.05) after ischemia. Endothelial progenitor cells also reduced inflammatory response after ischemia (p < 0.05). Endothelial progenitor cell transplantation reduced astrocyte-derived C3 expression in the brain after ischemic stroke, together with decreased C3aR and inflammatory response contributing to neurological function recovery. Our results indicate that modulating complement C3/C3aR pathway is a novel therapeutic target for the ischemic stroke.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Complement C3/metabolism , Endothelial Progenitor Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Astrocytes/metabolism , Atrophy/pathology , Brain/pathology , Brain Infarction/metabolism , Brain Injuries/pathology , Brain Ischemia/therapy , Case-Control Studies , Complement Activation/physiology , Disease Models, Animal , Endothelial Progenitor Cells/transplantation , Inflammation/metabolism , Male , Mice , Recovery of Function , Up-Regulation
5.
Am J Transl Res ; 11(5): 2830-2842, 2019.
Article in English | MEDLINE | ID: mdl-31217857

ABSTRACT

It has been shown that NF-κB signaling path is very effective pharmacological target for the treatment of various inflammatory diseases, including bacterial infection-associated acute kidney injury (AKI), which remains a main cause of disability and death in patients. Notably, IκB, the upstream molecular of NF-κB, plays an important role by inhibiting NF-κB activity, and IκB is regulated by cullin-RING E3 ligases (CRLs)-mediated proteasomal degradation. Therefore inhibition of CRLs-mediated neddylation and degradation of IκB would prevent NF-κB-mediated inflammation. MLN4924, a potent neddylation-inhibiting pharmacological agent, has been shown to have significant protective effects against lipopolysaccharide (LPS)-induced pro-inflammatory cytokine production through restriction of the CRL-mediated NF-κB pathway. However, it is still unclear whether MLN4924 plays a protective role through its anti-inflammatory properties in sepsis-induced AKI. In the current research, we explored whether MLN4924 have anti-inflammatory action in LPS-induced AKI mice. Our results show that MLN4924 dramatically decreased the cytotoxicity of LPS and inhibited LPS-induced synthesis and release of pro-inflammatory cytokines, such as TNF-α, IL-6 and IL-1ß, in HK2 cells, a renal tubular cell line. In addition, MLN4924 inhibited Nedd8-activating enzymes, which broke the process of cullin proteins neddylation and subsequent CRL target proteins degradation. The MLN4924-induced degradation of CRL attenuated the phosphorylation modification of IκB and IKK-α/ß and blocked the nuclear translocation of P50-NF-κB and P65-NF-κB in HK2 cells under LPS stimulation. Finally, our in vivo results show that MLN4924 protected against LPS-induced AKI at relatively low doses. Collectively, these results suggest that pharmacologically blocking neddylation by MLN4924 results in the suppression of pro-inflammatory cytokines generation through the CRL/NF-κB pathway in LPS-stimulated HK2 cells, and attenuated renal inflammation in LPS-induced AKI.

6.
Free Radic Res ; 53(7): 800-814, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31223046

ABSTRACT

Acute kidney injury (AKI) is a major kidney disease associated with poor clinical outcomes. Oxidative stress is predominantly involved in the pathogenesis of AKI. Autophagy and the Keap1-Nrf2 signalling pathway are both involved in the oxidative-stress response. However, the cross talk between these two pathways in AKI remains unknown. Here, we found that autophagy is upregulated during cisplatin-induced AKI. In contrast with previous studies, we observed a marked increase in p62. We also found that p62 knockdown reduces autophagosome formation and the expression of LC3II. To explore the cross talk between p62 and the Keap1-Nrf2 signalling pathway, HK-2 cells were transfected with siRNA targeting Nrf2, and we found that Nrf2 knockdown significantly reduced cisplatin-induced p62 expression. Moreover, p62 knockdown significantly decreased the protein expression of Nrf2, as well as Heme Oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase l (NQO1), whereas the expression of kelch-like ECH-associated protein 1 (Keap1) was upregulated. These results indicate that p62 creates a positive feedback loop in the Keap1-Nrf2 signalling pathway. Finally, we examined the role of p62 in cell protection during cisplatin-induced oxidative stress, and we found that p62 silencing in HK-2 cells increases apoptosis and reactive oxygen species (ROS) levels, which further indicates the protective role of p62 under oxidative stress and suggests that the cytoprotection 62 mediated is in part by regulating autophagic activity or the Keap1-Nrf2 signalling pathway. Taken together, our results have demonstrated a reciprocal regulation of p62, autophagy and the Keap1-Nrf2 signalling pathway under oxidative stress, which may be a potential therapeutic target against AKI.


Subject(s)
Acute Kidney Injury/metabolism , Cisplatin/toxicity , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Sequestosome-1 Protein/metabolism , Acute Kidney Injury/chemically induced , Animals , Autophagy , Humans , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Signal Transduction , Transfection
7.
Oncotarget ; 8(54): 92801-92814, 2017 Nov 03.
Article in English | MEDLINE | ID: mdl-29190957

ABSTRACT

The cellular hypoxia-reoxygenation (H/R) model is an ideal method to study ischemia-reperfusion injury, which is associated with high mortality. The role of microRNA-30c-5p (miR-30c-5p) in the H/R epithelial cell model remains unknown. In the current study, we observed a significant reduction in apoptosis when miR-30c-5p was up-regulated. We also found decreased levels of C-caspase-3 (C-CASP3) and Bcl-2-associated X (BAX) proteins and increased levels of B-cell lymphoma-2 (BCL2). Epidermal growth factor receptor (EGFR) showed similar results. Down-regulating miR-30c-5p increased the levels of apoptosis and C-CASP3 and BAX expression; additionally, cell proliferation was inhibited. Hypoxia-inducible factor 1α (HIF1α) protein expression levels were up-regulated in response to up-regulation of miR-30c-5p expression. The anti-apoptotic and proliferative effects of miR-30c-5p decreased significantly after the HIF1α protein levels were knocked down. Using a luciferase reporter assay, we confirmed that miR-30c-5p targets suppressor of cytokine signaling-3 (SOCS3). HIF1α levels increased when SOCS3 was blocked. Our data show that SOCS3 expression enhances apoptosis in the H/R model. In conclusion, up-regulating miR-30c-5p protects cells from H/R -induced apoptosis and induces cell proliferation; furthermore, HIF1α markedly contributes to this protective effect. MiR-30c-5p stabilizes HIF1α expression by targeting SOCS3 to achieve anti-apoptotic and proliferative effects.

9.
Stroke ; 48(12): 3375-3383, 2017 12.
Article in English | MEDLINE | ID: mdl-29146880

ABSTRACT

BACKGROUND AND PURPOSE: Striatal GABAergic neuron is known as a key regulator in adult neurogenesis. However, the specific role of striatal GABAergic neuronal activity in the promotion of neurological recovery after ischemic stroke remains unknown. Here, we used optogenetic approach to investigate these effects and mechanism. METHODS: Laser stimulation was delivered via an implanted optical fiber to inhibit or activate the striatal GABAergic neurons in Gad2-Arch-GFP or Gad2-ChR2-tdTomato mice (n=80) 1 week after 60-minute transient middle cerebral artery occlusion. Neurological severity score, brain atrophy volume, microvessel density, and cell morphological changes were examined using immunohistochemistry. Gene expression and protein levels of related growth factors were further examined using real-time polymerase chain reaction and Western blotting. RESULTS: Inhibiting striatal GABAergic neuronal activity improved functional recovery, reduced brain atrophy volume, and prohibited cell death compared with the control (P<0.05). Microvessel density and bFGF (basic fibroblast growth factor) expression in the inhibition group were also increased (P<0.05). In contrast, activation of striatal GABAergic neurons resulted in adverse effects compared with the control (P<0.05). Using cocultures of GABAergic neurons, astrocytes, and endothelial cells, we further demonstrated that the photoinhibition of GABAergic neuronal activity could upregulate bFGF expression in endothelial cells, depending on the presence of astrocytes. The conditioned medium from the aforementioned photoinhibited 3-cell coculture system protected cells from oxygen glucose deprivation injury. CONCLUSIONS: After ischemic stroke, optogenetic inhibition of GABAergic neurons upregulated bFGF expression by endothelial cells and promoted neurobehavioral recovery, possibly orchestrated by astrocytes. Optogenetically inhibiting neuronal activity provides a novel approach to promote neurological recovery.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/pathology , Corpus Striatum/metabolism , GABA Antagonists/therapeutic use , GABAergic Neurons/pathology , Optogenetics , Animals , Brain Ischemia/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/biosynthesis , Lasers , Male , Mice , Mice, Neurologic Mutants , Middle Cerebral Artery/pathology , Recovery of Function , gamma-Aminobutyric Acid/metabolism
10.
Exp Cell Res ; 360(2): 292-302, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28928081

ABSTRACT

Oxidative stress was predominantly involved in the pathogenesis of acute kidney injury (AKI). Recent studies had reported the protective role of specific microRNAs (miRNAs) against oxidative stress. Hence, we investigated the levels of miR140-5p and its functional role in the pathogenesis of Cisplatin induced AKI. A mice Cisplatin induced-AKI model was established. We found that miR-140-5p expression was markedly increased in mice kidney. Bioinformatics analysis revealed nuclear factor erythroid 2-related factor (Nrf2) was a potential target of miR-140-5p, We demonstrated that miR-140-5p did not affect Kelch-like ECH-associated protein 1 (Keap1) level but directly targeted the 3'-UTR of Nrf2 mRNA and played a positive role in the regulation of Nrf2 expression which was confirmed by luciferase activity assay and western blot. What was more, consistent with miR140-5p expression, the mRNA and protein levels of Nrf2, as well as antioxidant response element (ARE)-driven genes Heme Oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase l (NQO1) were significantly increased in mice kidney tissues. In vitro study, Enforced expression of miR-140-5p in HK2 cells significantly attenuated oxidative stress by decreasing ROS level and increasing the expression of manganese superoxide dismutase (MnSOD). Simultaneously, miR-140-5p decreased lactate dehydrogenase (LDH) leakage and improved cell vitality in HK2 cells under Cisplatin-induced oxidative stress. However, HK2 cells transfected with a siRNA targeting Nrf2 abrogated the protective effects of miR-140-5p against oxidative stress. These results indicated that miR-140-5p might exert its anti-oxidative stress function via targeting Nrf2. Our findings showed the novel transcriptional role of miR140-5p in the expression of Nrf2 and miR-140-5p protected against Cisplatin induced oxidative stress by activating Nrf2-dependent antioxidant pathway, providing a potentially therapeutic target in acute kidney injury.


Subject(s)
Acute Kidney Injury/chemically induced , Antioxidant Response Elements/drug effects , Cisplatin/pharmacology , Cytoprotection/genetics , MicroRNAs/physiology , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Animals , Antioxidant Response Elements/physiology , Cells, Cultured , Cytoprotection/drug effects , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1/physiology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...