Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Acta Biochim Biophys Sin (Shanghai) ; 56(3): 414-426, 2024 03 25.
Article in English | MEDLINE | ID: mdl-38429975

ABSTRACT

Diabetic nephropathy (DN) is a severe complication of diabetes and the leading cause of end-stage renal disease and death. Germacrone (Ger) possesses anti-inflammatory, antioxidant and anti-DN properties. However, it is unclear whether the improvement in kidney damage caused by Ger in DN mice is related to abnormal compositions and metabolites of the gut microbiota. This study generates a mouse model of DN to explore the potent therapeutic ability and mechanism of Ger in renal function by 16S rRNA sequencing and untargeted fecal metabolomics. Although there is no significant change in microbiota diversity, the structure of the gut microbiota in the DN group is quite different. Serratia_marcescens and Lactobacillus_iners are elevated in the model group but significantly decreased after Ger intervention ( P<0.05). Under the treatment of Ger, no significant differences in the diversity and richness of the gut microbiota are observed. An imbalance in the intestinal flora leads to the dysregulation of metabolites, and non-targeted metabolomics data indicate high expression of stearic acid in the DN group, and oleic acid could serve as a potential marker of the therapeutic role of Ger in the DN model. Overall, Ger improves kidney injury in diabetic mice, in part potentially by reducing the abundance of Serratia_marcescens and Lactobacillus_iners, as well as regulating the associated increase in metabolites such as oleic acid, lithocholic acid and the decrease in stearic acid. Our research expands the understanding of the relationship between the gut microbiota and metabolites in Ger-treated DN. This contributes to the usage of natural products as a therapeutic approach for the treatment of DN via microbiota regulation.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Lactobacillus , Animals , Mice , Diabetic Nephropathies/genetics , RNA, Ribosomal, 16S/genetics , Diabetes Mellitus, Experimental/genetics , Sesquiterpenes, Germacrane
3.
J Ethnopharmacol ; 325: 117820, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38286157

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Qingfei Xieding prescription was gradually refined and produced by Hangzhou Red Cross Hospital. The raw material includes Ephedra sinica Stapf, Morus alba L., Bombyx Batryticatus, Gypsum Fibrosum, Prunus armeniaca L. var. ansu Maxim., Houttuynia cordata Thunb. , Pueraria edulis Pamp. Paeonia L., Scutellaria baicalensis Georgi and Anemarrhena asphodeloides Bge. It is effective in clinical adjuvant treatment of patients with pulmonary diseases. AIM OF THE STUDY: To explore the efficacy and underlying mechanism of Qingfei Xieding (QF) in the treatment of bleomycin-induced mouse model. MATERIALS AND METHODS: TGF-ß induced fibrotic phenotype in vitro. Bleomycin injection induced lung tissue fibrosis mouse model in vivo. Flow cytometry was used to detect apoptosis, cellular ROS and lipid oxidation. Mitochondria substructure was observed by transmission electron microscopy. Autophagolysosome and nuclear entry of P65 were monitored by immunofluorescence. Quantitative real-time PCR was performed to detect the transcription of genes associated with mtDNA-cGAS-STING pathway and subsequent inflammatory signaling activation. RESULTS: TGF-ß induced the expression of α-SMA and Collagen I, inhibited cell viability in lung epithelial MLE-12 cells that was reversed by QF-containing serum. TGF-ß-mediated downregulation in autophagy, upregulation in lipid oxidation and ROS contents, and mitochondrial damage were rescued by QF-containing serum treatment, but CQ exposure, an autophagy inhibitor, prevented the protective role of QF. In addition to that, the decreased autophagolysosome in TGF-ß-exposed MLE-12 cells was reversed by QF and restored to low level in the combination treatment of QF and CQ. Mechanistically, QF-containing serum treatment significantly inhibited mtDNA-cGAS-STING pathway and subsequent inflammatory signaling in TGF-ß-challenged cells, which were abolished by CQ-mediated autophagy inhibition. In bleomycin-induced mouse model, QF ameliorated pulmonary fibrosis, reduced mortality, re-activated autophagy in lung tissues and restrained mtDNA-cGAS-STING inflammation pathway. However, the protective effects of QF in bleomycin-induced model mice were also abrogated by CQ. CONCLUSION: QF alleviated bleomycin-induced pulmonary fibrosis by activating autophagy, inhibiting mtDNA-cGAS-STING pathway-mediated inflammation. This research recognizes the protection role of QF on bleomycin-induced mouse model, and offers evidence for the potentiality of QF in clinical application for pulmonary fibrosis treatment.


Subject(s)
Pulmonary Fibrosis , Humans , Mice , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Bleomycin/toxicity , DNA, Mitochondrial/adverse effects , DNA, Mitochondrial/metabolism , Reactive Oxygen Species/metabolism , Lung , Transforming Growth Factor beta/metabolism , Mitochondria/metabolism , Inflammation/pathology , Disease Models, Animal , Autophagy , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/pharmacology , Nucleotidyltransferases/therapeutic use , Lipids/pharmacology , Transforming Growth Factor beta1/metabolism
4.
Int Immunopharmacol ; 126: 111316, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38056200

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a serious, lifelong lung disease with high morbidity and high mortality. Menstrual blood-derived stem cells (MenSCs) derived exosomes (MenSCs-Exo) emerge as an attractive tool for the treatment of acute lung injury and fibrosis-related diseases. However, more comprehensive mechanism over how MenSCs derived exosomes exhibits anti-pulmonary fibrosis needs to be elucidated. In this study, TGF-ß was used to construct cell fibrosis model, and bleomycin (BLM) was applied to induce lung tissue fibrosis mice model. BLM- and TGF-ß1-induced cellular reactive oxygen species (ROS), mitochondrial DNA (mtDNA) damage, and lung epithelial cell apoptosis were alleviated by MenSCs-Exo treatment in vivo and in vitro. Besides, it was found that MenSCs-Exo delivered miR-let-7 into MLE-12 cells/lung epithelial cell and the reduction of miR-let-7 blocked the improvement produced by MenSCs-Exo. Mechanistically, miR-let-7 directly bound to Sp3 and negatively regulated its expression. Sp3 elevation promoted the expression of ferroptosis-related protein and mitochondrial DNA (mtDNA) damage markers via recruiting HDAC2, thereby inactivating keap1/Nrf2 signal cascade, which were confirmed in BLM-induced pulmonary fibrosis mice model under the combination therapy of the MenSCs-Exo and let-7 inhibitor. Collectively, MenSCs derived exosomes could transmit miR-let-7 into MLE-12 cells to inhibit the expression of Sp3, thereby weakening the recruitment effect of Sp3 on HDAC2, lifting the deacetylation restriction of HDAC2 on Nrf2, and enhancing the Nrf2 pathway. These changes further declined ferroptosis and delayed the pathological process of oxidative damage and lung epithelial cell apoptosis in PF.


Subject(s)
Ferroptosis , Idiopathic Pulmonary Fibrosis , MicroRNAs , Mice , Animals , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Signal Transduction , Bleomycin/adverse effects , DNA, Mitochondrial/metabolism , Stem Cells/metabolism
5.
Free Radic Res ; 57(6-12): 413-429, 2023.
Article in English | MEDLINE | ID: mdl-37897414

ABSTRACT

Mitophagy is a critical intracellular event during the progression of diabetic nephropathy (DN). Our previous study demonstrated that germacrone has anti-ferroptotic properties and is a potential therapeutic agent for DN. However, the relationship among germacrone, mitophagy, and ferroptosis in DN remains unclear. In this study, the data confirmed that germacrone ameliorates high glucose (HG)-induced ferroptosis through limiting Fe (2+) content and lipid reactive oxygen species (ROS) accumulation in human kidney 2 (HK-2) cells. Germacrone reversed HG-mediated inhibition of mitophagy. Mitophagy inhibition and anabatic mitochondrial ROS abrogate germacrone-mediated protective effects against ferroptotic death, resulting in the subsequent activation of mitochondrial DNA (mtDNA) cytosolic leakage-induced stimulator of interferon response CGAMP interactor 1 (STING) signaling. The combination of a mitochondrial ROS antagonist and germacrone acts synergistically to alleviate the ferroptotic death of tubular cells and DN symptoms. In summary, germacrone ameliorated ferroptotic death in tubular cells by reactivating mitophagy and inhibiting mtDNA-STING signaling in DN. This study provides a novel insight into germacrone-mediated protection against DN progression and further confirms that antioxidant pharmacological strategies facilitate the treatment of DN.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Humans , Diabetic Nephropathies/drug therapy , Mitophagy , Reactive Oxygen Species/pharmacology , Kidney , DNA, Mitochondrial/pharmacology , DNA, Mitochondrial/therapeutic use
6.
Tuberculosis (Edinb) ; 143: 102424, 2023 12.
Article in English | MEDLINE | ID: mdl-37871493

ABSTRACT

BACKGROUND: The coexistence of pulmonary tuberculosis (PTB) and diabetes mellitus (DM) has emerged as a significant global public health concern. Patients with DM are at higher risk of developing PTB, and PTB is one of the important factors that exacerbate the development of DM. However, the impact of DM on the protein profile and underlying pathways in PTB patients is unclear. METHODS: We systematically used data-independent acquisition (DIA)-based liquid chromatography - tandem mass spectrometry (LC-MS/MS) to identify differentially expressed proteins (DEPs) in plasma samples from PTB patients, DM combined with PTB patients, and healthy controls. Then these DEPs were analyzed by bioinformatics. RESULTS: Our analysis identified 268 proteins, the results indicated that DEPs in the PTB group as well as in the DM-PTB group were mainly involved in immune responses, complement and coagulation cascade and cholesterol metabolic pathways compared to healthy controls. CONCLUSIONS: We analyzed the plasma protein profiles of PTB, DM-PTB, and HC groups using proteomics techniques and identified potential pathways for PTB patients with and without DM. This provides valuable clues to explore the impact of DM on PTB.


Subject(s)
Diabetes Mellitus , Mycobacterium tuberculosis , Tuberculosis, Pulmonary , Humans , Chromatography, Liquid , Proteomics , Tandem Mass Spectrometry , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/complications , Blood Proteins
7.
Microbiol Spectr ; 11(4): e0057723, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37522815

ABSTRACT

Pulmonary tuberculosis (PTB) and diabetes mellitus (DM) are common chronic diseases that threaten human health. Patients with DM are susceptible to PTB, an important factor that aggravates the complications of diabetes. However, the molecular regulatory mechanism underlying the susceptibility of patients with DM to PTB infection remains unknown. In this study, healthy subjects, patients with primary PTB, and patients with primary PTB complicated by DM were recruited according to inclusion and exclusion criteria. Peripheral whole blood was collected, and alteration profiles and potential molecular mechanisms were further analyzed using integrated bioinformatics analysis of metabolomics and transcriptomics. Transcriptional data revealed that lipocalin 2 (LCN2), defensin alpha 1 (DEFA1), peptidoglycan recognition protein 1 (PGLYRP1), and integrin subunit alpha 2b (ITGA2B) were significantly upregulated, while chloride intracellular channel 3 (CLIC3) was significantly downregulated in the group with PTB and DM (PTB_DM) in contrast to the healthy control (HC) group. Additionally, the interleukin 17 (IL-17), phosphatidylinositol 3-kinase (PI3K)-AKT, and peroxisome proliferator-activated receptor (PPAR) signaling pathways are important for PTB infection and regulation of PTB-complicated diabetes. Metabolomic data showed that glycerophospholipid metabolism, carbon metabolism, and fat digestion and absorption processes were enriched in the differential metabolic analysis. Finally, integrated analysis of both metabolomic and transcriptomic data indicated that the NOTCH1/JAK/STAT signaling pathway is important in PTB complicated by DM. In conclusion, PTB infection altered the transcriptional and metabolic profiles of patients with DM. Metabolomic and transcriptomic changes were highly correlated in PTB patients with DM. Peripheral metabolite levels may be used as biomarkers for PTB management in patients with DM. IMPORTANCE The comorbidity of diabetes mellitus (DM) significantly increases the risk of tuberculosis infection and adverse tuberculosis treatment outcomes. Most previous studies have focused on the relationship between the effect of blood glucose control and the outcome of antituberculosis treatment in pulmonary tuberculosis (PTB) with DM (PTB_DM); however, early prediction and the underlying molecular mechanism of susceptibility to PTB infection in patients with DM remain unclear. In this study, transcriptome sequencing and untargeted metabolomics were performed to elucidate the key molecules and signaling pathways involved in PTB infection and the susceptibility of patients with diabetes to PTB. Our findings contribute to the development of vital diagnostic biomarkers for PTB or PTB_DM and provide a comprehensive understanding of molecular regulation during disease progression.


Subject(s)
Diabetes Complications , Diabetes Mellitus , Tuberculosis, Pulmonary , Tuberculosis , Humans , Transcriptome , Phosphatidylinositol 3-Kinases , Tuberculosis, Pulmonary/genetics , Diabetes Mellitus/genetics , Biomarkers , Metabolomics , Metabolic Networks and Pathways/genetics
8.
Bioengineered ; 13(1): 774-788, 2022 01.
Article in English | MEDLINE | ID: mdl-34847832

ABSTRACT

Mitochondrial injury-triggered podocyte apoptosis is a major risk factor for diabetic nephropathy (DN). However, the detailed relationship between mitochondrial homeostasis and podocyte apoptosis remains unclear. The present study aimed to explore the role and functional mechanism of germacrone in DN in type I diabetes (type I DN). A mouse model of type I DN was established by injecting streptozocin, and a podocyte injury model was constructed using high glucose (HG) induction. Histopathology was detected by hematoxylin and eosin and periodic acid-Schiff staining. Transmission electron microscopy and flow cytometry were used to evaluate the mitochondrial function. Germacrone simultaneously reduced blood glucose, 24 h proteinuria, and other nephrotic symptoms in a type 1 DN mouse model. Moreover, germacrone protected against mitochondrial damage, limited reactive oxygen species (ROS) accumulation, and restored glutathione peroxidase (GPX) activity and GPX4 protein expression, subsequently preventing podocyte apoptosis. Mechanistically, the increased miR-188-3p expression in type I DN mice was reversed in germacrone-challenged DN mice. HG induced miR-188-3p expression and the miR-188-3p antagonist abolished the HG-mediated increase in ROS. Notably, miR-188-3p was found to have a therapeutic effect against DN by aggravating mitochondrial damage and podocyte apoptosis. Germacrone alleviates DN progression in type I diabetes by limiting podocyte apoptosis, which was partly counteracted by miR-188-3p upregulation. The combination of germacrone and miR-188-3p antagonists is expected to be an effective therapeutic strategy for DN.Abbreviations DN: diabetic nephropathy; Type I DN: DN in Type I diabetes; STZ: streptozocin; ROS: reactive oxygen species; NcRNAs: non-coding RNAs; UTR: untranslated regions; NC: negative control; BUN: blood urea nitrogen; BUA: blood uric acid; Ucr: urine creatinine; Scr: serum creatinine; PAS: Periodic Acid-Schiff; IF: Immunofluorescence; FISH: Fluorescence in situ hybridization; TUG1: taurine upregulated gene 1; GPX: Glutathione Peroxidase; GPX4: glutathione peroxidase 4; EMT: epithelial-mesenchymal transition.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/metabolism , MicroRNAs/metabolism , Mitochondria/metabolism , Podocytes/metabolism , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Male , Mice , MicroRNAs/genetics , Mitochondria/genetics , Mitochondria/pathology , Podocytes/pathology
9.
Antioxid Redox Signal ; 36(10-12): 740-759, 2022 04.
Article in English | MEDLINE | ID: mdl-34913724

ABSTRACT

Aims: Diabetic nephropathy (DN) is characterized by microalbuminuria, mainly associated with pathological and morphological alterations of podocyte. New drug targeting podocyte injury is a promising approach for treating DN. The present study is aimed at developing new drug targeting podocyte injury for treating DN. Results: In this study, germacrone ameliorated kidney damage and inhibited podocyte apoptosis in a DN mouse model. Based on RNA-seq, mmu_mmu_circRNA_0000309, located in host gene vascular endothelial zinc finger 1 (Vezf1), showed a sharp decline in DN mice and a remarkable recovery in germacrone-challenged DN mice. mmu_circRNA_0000309 silence or miR-188-3p mimics abrogated the antiapoptosis and anti-injury effects of germacrone through aggravating mitochondria damage, and elevating reactive oxygen species and ferroptosis-related protein levels. Mechanistically, mmu_circRNA_0000309 competitively sponged miR-188-3p, and subsequently promoted glutathione peroxidase 4 (GPX4) expression, thereby inactivating ferroptosis-dependent mitochondrial damage and podocyte apoptosis. In addition, GPX4 overexpression neutralized mmu_circRNA_0000309 silence-mediated mitochondria damage and ferroptosis in germacrone-exposed MPC5 cells. Innovation: We describe the novel effect and mechanism of germacrone on treating DN, which is linked to ferroptosis for the first time. Conclusion: mmu_circRNA_0000309 silence mediates drug resistance to germacrone in DN mice. mmu_circRNA_0000309 sponges miR-188-3p, and subsequently upregulates GPX4 expression, inactivating ferroptosis-dependent mitochondrial function and podocyte apoptosis. Possibly germacrone-based treatment for DN can be further motivated by regulating mmu_circRNA_0000309/miR-188-3p/GPX4 signaling axis. Antioxid. Redox Signal. 36, 740-759.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Ferroptosis , MicroRNAs , Animals , DNA-Binding Proteins , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Female , Ferroptosis/genetics , Humans , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , RNA, Circular/genetics , Sesquiterpenes, Germacrane , Transcription Factors
11.
Oxid Med Cell Longev ; 2021: 6634601, 2021.
Article in English | MEDLINE | ID: mdl-34257814

ABSTRACT

Oxidative stress (OxS) is a wildly described cause of damage to macromolecules, resulting in abnormal physiological conditions. In recent years, a few studies have shown that oxidation/antioxidation imbalance plays a significant role in developing diseases involving different systems and organs. However, the research on the circular RNA (circRNA) roles in OxS is still in its very infancy. Therefore, we hope to provide a comprehensive overview of the recent research that explored the function of circRNAs associated with OxS and its role in the pathogenesis of different diseases that affect different body systems like the nervous system, cardiovascular system, kidneys, and lungs. It provides the possibilities of using these circRNAs as superior diagnostic and therapeutic options for OxS associated with these disease conditions.


Subject(s)
Biomarkers/metabolism , Oxidative Stress/genetics , RNA, Circular/genetics , Humans
12.
Front Cell Infect Microbiol ; 11: 783049, 2021.
Article in English | MEDLINE | ID: mdl-35111693

ABSTRACT

BACKGROUND: Gut-microbiota-brain axis links the relationship between intestinal microbiota and sepsis-associated encephalopathy (SAE). However, the key mediators between them remain unclear. METHODS: Memory test was determined by Water maze. Intestinal flora was measured by 16S RNA sequencing. Neurotransmitter was detected by high-performance liquid chromatography (HPLC). Histopathology was determined by H&E, immunofluorescence (IF), and terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) staining. Flow cytometry was employed to determine the proportion of macrophages. RESULTS: Fecal microbiota transplantation (FMT) relieved hippocampus impairment of SAE rats by inhibiting inflammation cytokine secretion, the expression of IBA-1 and neurotransmitter disturbance, and cell apoptosis and autophagy, accompanied by the reduced M1 polarization and M1 pro-inflammation factors produced by macrophages in mesenteric lymph nodes (MLNs). Actually, M1 polarization in SAE rats depended on intestinal epithelial cell (IEC)-derived exosome. GW4869-initiated inhibition of exosome secretion notably abolished M1 polarization and the secretion of IL-1ß. However, GW4869-mediated improvement of hippocampus impairment was counteracted by the delivery of recombinant interleukin (IL)-1ß to hippocampus. Mechanistically, IEC-derived exosome induced the excessive circulating IL-1ß produced by CP-R048 macrophages, which subsequently induced damage and apoptosis of hippocampal neurons H19-7 in an autophagy-dependent manner. And reactivation of autophagy facilitates intestinal IL-1ß-mediated hippocampal neuron injury. CONCLUSION: Collectively, intestinal flora disturbance induced the exosome release of IECs, which subsequently caused M1 polarization in MLNs and the accumulation of circulating IL-1ß. Circulating IL-1ß promoted the damage and apoptosis of neurons in an autophagy-dependent manner. Possibly, targeting intestinal flora or IEC-derived exosome contributes to the treatment of SAE.


Subject(s)
Exosomes , Sepsis-Associated Encephalopathy , Animals , Epithelial Cells , Fecal Microbiota Transplantation , Neurons , Rats
13.
Mol Med Rep ; 22(4): 2741-2752, 2020 10.
Article in English | MEDLINE | ID: mdl-32945378

ABSTRACT

Gallium (Ga) ions have been widely utilized for biomedical applications; however, their role in osteoblast regulation is not completely understood. The aim of the present study was to investigate the potential effect of Ga ions on osteoinduction in two osteoblast cell lines and to explore the underlying mechanisms. Human hFOB1.19 and mouse MC3T3­E1 osteoblasts were treated with Ga nitride (GaN) at different concentrations, and the degree of osteoinduction was assessed. Ga ion treatment was found to increase alkaline phosphatase activity and accelerate calcium nodule formation, as assessed using ALP activity assay and Alizarin red S staining. Moreover, upregulated expression levels of osteogenic proteins in osteoblasts were identified using western blotting and reverse transcription­quantitative PCR. Western blotting was also performed to demonstrate that the biological action of Ga ions was closely associated with the transient receptor potential melastatin 7/Akt signaling pathway. Furthermore, it was found that Ga ions did not cause osteoblast apoptosis, as indicated via flow cytometry, but promoted osteoclast proliferation, migration or invasion. The present study investigated the potential role of Ga ions in regulating osteoinduction of osteoblasts, thereby providing a promising strategy for the treatment of osteoporosis.


Subject(s)
Gallium/pharmacology , Osteoblasts/cytology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TRPM Cation Channels/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Humans , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , TRPM Cation Channels/genetics
14.
Biomark Med ; 14(10): 839-854, 2020 07.
Article in English | MEDLINE | ID: mdl-32856461

ABSTRACT

Background: Proteome studies for multiple renal diseases is bare. Methodology & results: Using isobaric tags for relative and absolute quantitation labeling, many differentially expressed proteins (DEPs) were identified in acute kidney injury (AKI), AKI + chronic kidney disease (CKD), diabetic CKD and nondiabetic CKD with or without IgA nephropathy (IgAN). Comparative analysis indicated that 34, 35, 17, 91 and 14 unique DEPs were found in AKI, AKI + CKD, CKD, diabetic CKD and nondiabetic CKD. Compared with nondiabetic CKD with IgAN, 47 unique DEPs were found in that without IgAN. Serum amyloid A1 (SAA1) and hepatocyte growth factor activator were unregulated in AKI and nondiabetic CKD without IgAN, respectively. Regenerating islet-derived protein 3-α (Reg3A) upregulation is associated with AKI and AKI + CKD patients. Conclusion: This research contributes to urinary biomarker discovery from multiple renal diseases.


Subject(s)
Biomarkers/urine , Kidney Diseases/urine , Proteomics , Adult , Biomarkers/metabolism , Female , Humans , Kidney Diseases/metabolism , Male
16.
Biomed Res Int ; 2020: 2685305, 2020.
Article in English | MEDLINE | ID: mdl-32149094

ABSTRACT

BACKGROUND: Podocyte migration is actively involved in the process of podocyte loss and proteinuria production, which is closely associated with the development of diabetic nephropathy (DN). Exosomes from adipose-derived stem cells (ADSCs-Exos) effectively inhibit podocyte apoptosis in the treatment of DN. However, how ADSCs-Exos affect the migration of podocytes is obscure. This study is aimed at exploring the regulatory role of ADSCs-Exos on cell migration and the underlying mechanism. METHODS: ADSCs-Exo was authenticated by transmission electron microscopy (TEM), western blotting, and flow cytometry. Cell viability and migration ability of podocytes were measured by CCK8 and Transwell assays, respectively. Relative expressions of miRNAs and mRNAs were determined by qRT-PCR. The transmitting between PKH26-labeled exosome and podocytes was evaluated by IF assay. Dual luciferase reporter assay was employed to detect the relationship between miR-215-5p and ZEB2. RESULTS: The exposure to serum from DN patient (hDN-serum) significantly inhibited cell viability of podocytes, but ADSCs-Exo addition notably blunts cytotoxicity induced by the transient stimulus of hDN-serum. Besides, ADSCs-Exo administration powerfully impeded high glucose- (HG-) induced migration and injury of podocyte. With the podocyte dysfunction, several miRNAs presented a significant decline under the treatment of HG including miR-251-5p, miR-879-5p, miR-3066-5p, and miR-7a-5p, all of which were rescued by the addition of ADSCs-Exo. However, only miR-251-5p was a key determinant in the process of ADSCs-Exo-mediated protective role on podocyte damage. The miR-251-5p inhibitor counteracted the improvement from the ADSCs-Exo preparation on HG-induced proliferation inhibition and migration promotion. Additionally, miR-215-5p mimics alone remarkably reversed HG-induced EMT process of podocyte. Mechanistically, we confirmed that ADSCs-Exos mediated the shuttling of miR-215-5p to podocyte, thereby protecting against HG-induced metastasis, possibly through inhibiting the transcription of ZEB2. CONCLUSION: ADSCs-Exo has the protective effect on HG-evoked EMT progression of podocytes thru a mechanism involving ZEB2. Potentially, the ADSCs-Exo preparation is a useful therapeutic strategy for improving podocyte dysfunction and DN symptoms clinically.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs , Zinc Finger E-box Binding Homeobox 2/antagonists & inhibitors , Animals , Cell Line , Cell Survival/drug effects , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/pharmacology , Podocytes/drug effects , Zinc Finger E-box Binding Homeobox 2/metabolism
17.
Can Respir J ; 2019: 7930396, 2019.
Article in English | MEDLINE | ID: mdl-31781316

ABSTRACT

Background: Imbalanced oxidative stress and antioxidant defense are involved in airway remodeling in asthma. It has been demonstrated that Tetrandrine has a potent role in antioxidant defense in rheumatoid arthritis and hypertension. However, the correlation between Tetrandrine and oxidative stress in asthma is utterly blurry. This study aimed to investigate the role of Tetrandrine on oxidative stress-mediated airway remolding. Materials and Methods: Chronic asthma was established by ovalbumin (OVA) administration in male Wistar rats. Histopathology was determined by HE staining. Immunofluorescence was employed to detect the expression of α-SMA and Nrf-2. Level of oxidative stress and matrix metalloproteinases were examined by ELISA kits. Cell viability and cell cycle of primary airway smooth muscle cells (ASMCs) were evaluated by CCK8 and flow cytometry, respectively. Signal molecules were detected using western blot. Results: Tetrandrine effectively impairs OVA-induced airway inflammatory and airway remodeling by inhibiting the expression of CysLT1 and CysLTR1. The increase of oxidative stress and subsequent enhancement of MMP9 and TGF-ß1 expression were rescued by the administration of Tetrandrine in the rat model of asthma. In in vitro experiments, Tetrandrine markedly suppressed TGF-ß1-evoked cell viability and cell cycle promotion of ASMCs in a dose-dependent manner. Furthermore, Tetrandrine promoted Nrf-2 nuclear transcription and activated its downstream HO-1 in vivo and in vitro. Conclusion: Tetrandrine attenuates airway inflammatory and airway remodeling in rat model of asthma and TGF-ß1-induced cell proliferation of ASMCs by regulating oxidative stress in primary ASMCs, suggesting that Tetrandrine possibly is an effective candidate therapy for asthma.


Subject(s)
Airway Remodeling/drug effects , Asthma/drug therapy , Benzylisoquinolines/therapeutic use , Immunosuppressive Agents/therapeutic use , Animals , Asthma/complications , Asthma/metabolism , Benzylisoquinolines/pharmacology , Chronic Disease , Disease Models, Animal , Drug Evaluation, Preclinical , Heme Oxygenase-1/metabolism , Immunosuppressive Agents/pharmacology , Male , Matrix Metalloproteinases/metabolism , Membrane Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Random Allocation , Rats, Wistar , Receptors, Leukotriene/metabolism
18.
Oxid Med Cell Longev ; 2019: 4506303, 2019.
Article in English | MEDLINE | ID: mdl-31949877

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a prototype of chronic, progressive, and fibrotic lung disease with high morbidity and high mortality. Menstrual blood-derived stem cells (MenSCs) have proven to be an attractive tool for the treatment of acute lung injury and fibrosis-related diseases through immunosuppression and antifibrosis. However, whether MenSC-derived exosomes have the similar function on pulmonary fibrosis remains unclear. In the present study, exosomes secreted from MenSCs (MenSCs-Exo) were verified by transmission electron microscope (TEM), nanoparticle tracking analyzer (NTA), and western blotting. And MenSC-Exo addition significantly improved BLM-induced lung fibrosis and alveolar epithelial cell damage in mice, mainly reflected in BLM-mediated enhancement of the fibrosis score, blue collagen deposition, dry/wet gravity ratio, hydroxyproline and malondialdehyde levels, and downregulation of glutathione peroxidase, which were all robustly reversed by MenSC-Exo management. Additionally, BLM- and TGF-ß1-evoked cellular reactive oxygen species (ROS), mitochondrial DNA (mtDNA) damage, and cell apoptosis were rescued by MenSCs-Exo in vivo and in vitro. Further study indicated that the MenSCs-Exo could transport miRNA Let-7 into recipient alveolar epithelial cells. Let-7 inhibitor administration significantly blocked the exosome-mediated improvement role on lung fibrosis in mice. Mechanistically, Let-7 was able to regulate the expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX1) through binding to its 3'-UTR region. Forced expression of LOX1 promoted the expression of apoptosis-related protein and mtDNA damage markers via regulating NLRP3 which was also confirmed in BLM model mice under the combination therapy of the exosome and Let-7 inhibitor. Collectively, this study demonstrates that exosomal Let-7 from MenSCs remits pulmonary fibrosis through regulating ROS, mtDNA damage, and NLRP3 inflammasome activation. This provides a new approach of exocytosis on the treatment of fibrotic lung disease.


Subject(s)
DNA Damage , Endometrium/cytology , Exosomes/genetics , MicroRNAs/genetics , Mitochondria/metabolism , Pulmonary Fibrosis/therapy , Stem Cell Transplantation , Stem Cells/cytology , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis , Bleomycin/toxicity , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Disease Models, Animal , Female , Humans , Male , Menstruation/blood , Mice , Mice, Inbred C57BL , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Reactive Oxygen Species , Scavenger Receptors, Class E/metabolism , Transforming Growth Factor beta1/metabolism
19.
Clin Sci (Lond) ; 132(20): 2175-2188, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30232174

ABSTRACT

Increased production of reactive oxygen species (ROS) and inflammation are major contributors to the development and progression of diabetes-associated erectile dysfunction (DMED). As an endogenous antioxidant and anti-inflammatory factor, the potential implication of pigment epithelium-derived factor (PEDF) in DMED has not been revealed. To assess the potential antioxidant and anti-inflammatory functions of PEDF in DMED, we first demonstrated that PEDF was significantly decreased at the levels of the mRNA and protein in the penis of diabetic rats compared with normal controls. To test the hypothesis that decreased the penile levels of PEDF are associated with oxidative stress and inflammation in DMED, an adenovirus expressing PEDF (Ad-PEDF) or the same titer of control virus (Ad-GFP) was intracavernously administered at 2 weeks after diabetic onset. After 6 weeks of treatment, we found that administration of Ad-PEDF could significantly increase erectile response to cavernosal nerve stimulation in the diabetic rats by restoring the endothelial NO synthase (eNOS), P-eNOS, and neuronal NO synthase (nNOS) protein levels to the standard levels represented in normal rats and by suppressing the levels of tumor necrosis factor-α (TNF-α) and oxidative stress. In conclusion, the present data indicated that the antioxidant and anti-inflammatory potential of PEDF plays important role in restoring erectile function by the inhibition of oxidative stress and TNF-α production.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Eye Proteins/genetics , Nerve Growth Factors/genetics , Penile Erection/genetics , Penis/metabolism , Serpins/genetics , Animals , Diabetes Mellitus, Experimental/metabolism , Down-Regulation , Eye Proteins/metabolism , Gene Expression Regulation , Male , Nerve Growth Factors/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Serpins/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
J Biomed Nanotechnol ; 14(1): 98-113, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29463368

ABSTRACT

(Arginine-Glycine-Aspartic)-methoxy polyethylene glycol-(1,2-distearoyl-sn-glycero-3-phosphoethanolaMine-N) (abbreviation: RGD-PEG2000-DSPE or RGD-PD) was successfully synthesized and verified by 1H-NMR and MALDI-TOF MS. Polyethylene glycol-poly-L-lysine/RGD-PD/phospholipid/calcium phosphate nanoparticles (PEG-PLL/RGD-PD/PL/CaP NPs or MNPs) were prepared using a novel, simple method conducted at room temperature. Transmission electron microscopy (TEM) analysis showed that the MNPs were spheres of uniform size, with a diameter of ∼30 nm, and smooth surface. Thermogravimetric analysis (TGA) revealed that the PEG-PLL/RGD-PD/PL micelle was packed in the CaP shell. MNPs had little effect on hemolysis, coagulation, cardiac oxidative stress, inflammatory response and DNA damage, indicating negligible cytotoxicity in vitro and in vivo. Experiments in Zebrafish indicated that the MNPs neither affected the survival rate and heartbeat rate, nor induced malformation and apoptosis during embryogenesis. In conclusion, these results demonstrate that the newly-developed MNPs have good biocompatibility and a great potential as drug and gene carrier.


Subject(s)
Nanoparticles , Phosphatidylethanolamines , Polyethylene Glycols , Materials Testing , Phospholipids
SELECTION OF CITATIONS
SEARCH DETAIL
...