Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(22)2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36430348

ABSTRACT

Cancers of the oral cavity can develop in the anatomic area extending from the lip, gum, tongue, mouth, and to the palate. Histologically, about 85-90% of oral cavity cancers are of the type squamous cells carcinomas (SCCs). The incidence of oral tongue SCC is higher in the tongue than any other anatomic area of the oral cavity. Here, we investigated the therapeutic effects and molecular mechanisms of docetaxel, which is a paclitaxel antitumor agent, on the cell growth of a human tongue SCC-derived SAS cell line. The results showed that docetaxel (10-300 nM) induced cytotoxicity and caspase-3 activity in SAS cells. Moreover, docetaxel (100 nM) promoted the expression of apoptosis-related signaling molecules, including the cleavages of caspase-3, caspase-7, and poly (ADP-ribose) polymerase (PARP). In mitochondria, docetaxel (100 nM) decreased the mitochondrial membrane potential (MMP) and Bcl-2 mRNA and protein expression and increased cytosolic cytochrome c protein expression and Bax mRNA and protein expression. In terms of mitogen-activated protein kinase (MAPK) and adenosine monophosphate-activated protein kinase (AMPK) signaling, docetaxel increased the expression of phosphorylated (p)-extracellular signal-regulated kinase (ERK), p-c-Jun N-terminal kinase (JNK), and p-AMPKα protein expression but not p-p38 protein expression. Moreover, the increase in caspase-3/-7 activity and Bax protein expression and decreased Bcl-2 protein expression and MMP depolarization observed in docetaxel-treated SAS cells could be reversed by treatment with either SP600125 (a JNK inhibitor), PD98059 (an MEK1/2 (mitogen-activated protein kinase kinase 1/2) inhibitor), or compound c (an AMPK inhibitor). The docetaxel-induced increases in p-JNK, p-ERK, and p-AMPKα protein expression could also be reversed by treatment with either SP600125, PD98059, or compound c. These results indicate that docetaxel induces human tongue SCC cell apoptosis via interdependent MAPK-JNK, MAPK-ERK1/2, and AMPKα signaling pathways. Our results show that docetaxel could possibly exert a potent pharmacological effect on human oral tongue SCC cell growth.


Subject(s)
Carcinoma, Squamous Cell , Tongue Neoplasms , Humans , Extracellular Signal-Regulated MAP Kinases/metabolism , Docetaxel/pharmacology , Caspase 3/metabolism , AMP-Activated Protein Kinases , Carcinoma, Squamous Cell/drug therapy , Tongue Neoplasms/drug therapy , Apoptosis , Proto-Oncogene Proteins c-bcl-2 , Epithelial Cells/metabolism , Tongue/metabolism , RNA, Messenger
2.
Molecules ; 27(18)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36144517

ABSTRACT

An ischemic stroke is brain damage caused by interruption of blood supply to the brain that can cause death and long-term disability. New medical strategies or therapies are urgently needed for ischemic stroke. Icaritin (ICT) is a metabolite of icariin (ICA), which are two active flavonoid components extracted from Herba epimedii and considered neuroprotective agents in animal models of Alzheimer's disease and ischemic stroke. The therapeutic effect of ICT on ischemic still remains to be clarified. The aim of this study was to investigate the therapeutic effect of ICT on cerebral ischemia-reperfusion-associated senescence and apoptosis in a middle cerebral artery occlusion (MCAO) mouse model (ischemia for 50 min and reperfusion for 24 h). Administration of ICT after ischemia significantly reduced MCAO-induced neurological damage, infarct volume, and histopathological changes in the brain of acute ischemic stroke mice. ICT treatment could also reduce neuronal apoptosis and senescence and reversed the expression of apoptosis- and senescence-related signaling proteins. These findings suggest that ICT may have therapeutic potential to ameliorate acute ischemic stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Neuroprotective Agents , Reperfusion Injury , Stroke , Animals , Apoptosis , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Disease Models, Animal , Flavonoids/pharmacology , Flavonoids/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Reperfusion , Reperfusion Injury/metabolism , Stroke/drug therapy
3.
Biochem Biophys Res Commun ; 608: 122-127, 2022 06 11.
Article in English | MEDLINE | ID: mdl-35397424

ABSTRACT

Acute kidney injury (AKI) is known as a sudden episode of kidney injury, which happens suddenly within a few hours or a few days. Quercetin (3,3',4',5,7-pentahydroxyflavone) is a flavonoid found in plants. Quercetin is known to have several biological activities, such as anti-oxidant, anti-inflammatory, and anti-carcinogenic effects. However, low water solubility and bioavailability are the limitations of quercetin for its clinical applications. Moreover, ischemia/reperfusion (I/R) injury is a common cause of AKI. There are no satisfactory strategies for I/R-induced AKI. Developing suitable preventive or therapeutic intervention for AKI is an important and urgent issue. We investigated the benefit effect of synthesized polyethylene glycol (PEG) conjugated polyethyleneimine (PEI) nanoparticles for targeted delivery of quercetin on AKI in a mouse model. An I/R-induced AKI mouse model was used to evaluate the therapeutic effect of quercetin polymeric nanoparticles by intravenous injection. Biochemical changes for renal function in blood samples were analyzed. Histological and immunohistochemical changes were also analyzed. The biochemical changes of blood urea nitrogen (BUN), creatinine, and cystatin C were significantly increased in I/R-induced AKI mice, which could be significantly reversed by quercetin polymeric nanoparticles. Quercetin polymeric nanoparticles could also significantly decrease the histological lesions, positive staining for 3-nitrotyrosine and cyclooxygenase-2, and lipid peroxidation in the kidneys of I/R-induced AKI mice. These results demonstrate for the first time that quercetin polymeric nanoparticles possess therapeutic potential for the treatment of I/R-induced AKI in vivo.


Subject(s)
Acute Kidney Injury , Nanoparticles , Reperfusion Injury , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Animals , Disease Models, Animal , Female , Humans , Ischemia/pathology , Kidney/pathology , Male , Mice , Quercetin/pharmacology , Reperfusion , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology
4.
Biomedicines ; 10(3)2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35327397

ABSTRACT

Nobiletin (Nob), a critical active flavonoid of citrus fruits, has received attention for its superior physical functions, which have shown to improve the progression of diseases. Chronic kidney disease (CKD) is recognized as a global health problem, and its mortality and morbidity rates are worsened with an increased risk of accompanying disorders. In this study, we aimed to elucidate whether Nob treatment ameliorates kidney fibrosis and also to identify the potential signaling networks in a unilateral ureteral obstructive (UUO) mouse model, which was used to mimic the progression of CKD. Six-week-old C57BL/6J mice were orally treated with 50 mg/kg of Nob for 14 constitutive days after UUO surgery. We found that the administration of Nob diminished kidney fibrosis and the expression of EMT markers, ameliorated oxidative stress and ferroptosis-associated injury, and mitigated the inflammatory response in the kidneys of UUO mice. Our results suggested that Nob treatment has antiferroptosis, anti-inflammatory, and antifibrotic effects, improving the progression of CKD in UUO mice. Nob may serve as a potential therapeutic candidate for the improvement of progressive CKD in further studies.

5.
Int J Mol Sci ; 23(5)2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35270009

ABSTRACT

Methylmercury (MeHg), a long-lasting organic pollutant, is known to induce cytotoxic effects in mammalian cells. Epidemiological studies have suggested that environmental exposure to MeHg is linked to the development of diabetes mellitus (DM). The exact molecular mechanism of MeHg-induced pancreatic ß-cell cytotoxicity is still unclear. Here, we found that MeHg (1-4 µM) significantly decreased insulin secretion and cell viability in pancreatic ß-cell-derived RIN-m5F cells. A concomitant elevation of mitochondrial-dependent apoptotic events was observed, including decreased mitochondrial membrane potential and increased proapoptotic (Bax, Bak, p53)/antiapoptotic (Bcl-2) mRNA ratio, cytochrome c release, annexin V-Cy3 binding, caspase-3 activity, and caspase-3/-7/-9 activation. Exposure of RIN-m5F cells to MeHg (2 µM) also induced protein expression of endoplasmic reticulum (ER) stress-related signaling molecules, including C/EBP homologous protein (CHOP), X-box binding protein (XBP-1), and caspase-12. Pretreatment with 4-phenylbutyric acid (4-PBA; an ER stress inhibitor) and specific siRNAs for CHOP and XBP-1 significantly inhibited their expression and caspase-3/-12 activation in MeHg-exposed RIN-mF cells. MeHg could also evoke c-Jun N-terminal kinase (JNK) activation and reactive oxygen species (ROS) generation. Antioxidant N-acetylcysteine (NAC; 1mM) or 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (trolox; 100 µM) markedly prevented MeH-induced ROS generation and decreased cell viability in RIN-m5F cells. Furthermore, pretreatment of cells with SP600125 (JNK inhibitor; 10 µM) or NAC (1 mM) or transfection with JNK-specific siRNA obviously attenuated the MeHg-induced JNK phosphorylation, CHOP and XBP-1 protein expression, apoptotic events, and insulin secretion dysfunction. NAC significantly inhibited MeHg-activated JNK signaling, but SP600125 could not effectively reduce MeHg-induced ROS generation. Collectively, these findings demonstrate that the induction of ROS-activated JNK signaling is a crucial mechanism underlying MeHg-induced mitochondria- and ER stress-dependent apoptosis, ultimately leading to ß-cell death.


Subject(s)
Endoplasmic Reticulum Stress , Methylmercury Compounds , Animals , Apoptosis , Caspase 3/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Mammals/metabolism , Methylmercury Compounds/pharmacology , Mitochondria/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
6.
Cancers (Basel) ; 13(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34885061

ABSTRACT

CARD-recruited membrane-associated protein 3 (CARMA3) is overexpressed in various cancers and is associated with cancer cell proliferation, metastasis, and tumor progression; however, the underlying mechanisms of CARMA3 in colorectal cancer (CRC) metastasis remain unclear. Here, we found that higher CARMA3 expression was correlated with poor overall survival and metastasis in CRC patients from the TNMplot database and Human Tissue Microarray staining. Elevating CARMA3 expression promoted cell proliferation, epithelial-mesenchymal transition (EMT) induction, migration/invasion abilities, sphere formation, and cancer stem cell markers expression. Knockdown of CARMA3 decreased these processes via the EMT-related transcription factor Slug. Moreover, CARMA3 depletion significantly reduced tumor growth in mice that were consistent with the in vitro results. CRC migration/invasion could be regulated by CARMA3/YAP/Slug signaling axis using genetic inhibition of Yes-associated protein (YAP). Interestingly, CARMA3 induced activation of nuclear factor (NF)-κB through YAP expression, contributing to upregulation of Slug. YAP expression positively correlated with CARMA3, NF-κB, and Slug gene expression and poor clinical outcomes in CRC patients. Our findings demonstrate for the first time that CARMA3 plays an important role in CRC progression, which may serve as a potential diagnostic biomarker and candidate therapeutic target for CRC treatment.

7.
Biomedicines ; 9(11)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34829948

ABSTRACT

Stroke, which is the second leading cause of mortality in the world, is urgently needed to explore the medical strategies for ischemic stroke treatment. Both icariin (ICA) and icaritin (ICT) are the major active flavonoids extracted from Herba epimedii that have been regarded as the neuroprotective agents in disease models. In this study, we aimed to investigate and compare the neuroprotective effects of ICA and ICT in a middle cerebral artery occlusion (MCAO) mouse model. Male ICR mice were pretreated with both ICA and ICT, which ameliorated body weight loss, neurological injury, infarct volume, and pathological change in acute ischemic stroke mice. Furthermore, administration of both ICA and ICT could also protect against neuronal cell apoptotic death, oxidative and nitrosative stress, lipid peroxidation, and extracellular matrix (ECM) accumulation in the brains. The neuroprotective effects of ICT are slightly better than that of ICA in acute cerebral ischemic stroke mice. These results suggest that pretreatment with both ICA and ICT improves the neuronal cell apoptosis and responses of oxidative/nitrosative stress and counteracts the ECM accumulation in the brains of acute cerebral ischemic stroke mice. Both ICA and ICT treatment may serve as a useful therapeutic strategy for acute ischemic stroke.

8.
Nutrients ; 13(11)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34836176

ABSTRACT

Nootkatone is one of the major active ingredients of Alpiniae oxyphyllae, which has been used as both food and medicinal plants for the treatment of diarrhea, ulceration, and enuresis. In this study, we aimed to investigate whether nootkatone treatment ameliorated the progression of chronic kidney diseases (CKD) and clarified its underlying mechanisms in an obstructive nephropathy (unilateral ureteral obstructive; UUO) mouse model. Our results revealed that nootkatone treatment preventively decreased the pathological changes and significantly mitigated the collagen deposition as well as the protein expression of fibrotic markers. Nootkatone could also alleviate oxidative stress-induced injury, inflammatory cell infiltration, and renal cell apoptotic death in the kidneys of UUO mice. These results demonstrated for the first time that nootkatone protected against the progression of CKD in a UUO mouse model. It may serve as a potential therapeutic candidate for CKD intervention.


Subject(s)
Apoptosis/drug effects , Inflammation/drug therapy , Polycyclic Sesquiterpenes/pharmacology , Renal Insufficiency, Chronic/drug therapy , Alpinia/chemistry , Animals , Disease Models, Animal , Fibrosis/drug therapy , Fibrosis/metabolism , Inflammation/metabolism , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Renal Insufficiency, Chronic/metabolism
9.
Eur J Pharm Biopharm ; 168: 38-52, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34450241

ABSTRACT

Tumor hypoxia is a common feature in colorectal cancer (CRC), and is associated with resistance to radiotherapy and chemotherapy. Thus, a specifically targeted probe for the detection of hypoxic CRC cells is urgently needed. Carbonic anhydrase 9 (CA9) is considered to be a specific marker for hypoxic CRC diagnosis. Here, a nuclear imaging Indium-111 (111In)-labeled dual CA9-targeted probe was synthesized and evaluated for CA9 detection in in vitro, in vivo, and in human samples. The CA9-targeted peptide (CA9tp) and CA9 inhibitor acetazolamide (AAZ) were combined to form a dual CA9-targeted probe (AAZ-CA9tp) using an automatic microwave peptide synthesizer, which then was conjugated with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) for radioisotope (111In) labeling (111In-DOTA-AAZ-CA9tp). The assays for cell binding, stability, and toxicity were conducted in hypoxic CRC HCT15 cells. The analyses for imaging and biodistribution were performed in an HCT15 xenograft mouse model. The binding and distribution of 111In-DOTA-AAZ-CA9tp were detected in human CRC samples using microautoradiography. AAZ-CA9tp possessed good CA9-targeting ability in hypoxic HCT15 cells. The dual CA9-targeted radiotracer showed high serum stability, high surface binding, and high affinity in vitro. After exposure of 111In-DOTA-AAZ-CA9tp to the HCT15-bearing xenograft mice, the levels of 111In-DOTA-AAZ-CA9tp were markedly and specifically increased in the hypoxic tumor tissues compared to control mice. 111In-DOTA-AAZ-CA9tp also targeted the areas of CA9 overexpression in human colorectal tumor tissue sections. The results of this study suggest that the novel 111In-DOTA-AAZ-CA9tp nuclear imaging agent may be a useful tool for the detection of hypoxic CRC cells in clinical practice.


Subject(s)
Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/metabolism , Colorectal Neoplasms/diagnostic imaging , Tomography, Emission-Computed, Single-Photon/methods , Acetazolamide/pharmacology , Animals , Carbonic Anhydrase Inhibitors/pharmacology , Cell Hypoxia , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Indium Radioisotopes , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Tissue Distribution , Xenograft Model Antitumor Assays
10.
Sci Rep ; 10(1): 14317, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32868865

ABSTRACT

The therapeutic effects of low intensity pulsed ultrasound (LIPUS) on renal ischemia/reperfusion injury (IRI) with acute kidney injury (AKI) are still unclear. A renal tubule cell model under H2O2 or hypoxia/reoxygenation (H/R) conditions with or without LIPUS pre-treatment (1 MHz, 30 and 100 mW/cm2, 15 min) was used to test the in vitro effects of LIPUS. An AKI mouse model of unilateral IRI with nephrectomy of the contralateral kidney for 48 h with or without LIPUS treatment (3 MHz, 100 mW/cm2, 20 min/day) 5 day before IRI were used to investigate the in vivo effects of LIPUS. LIPUS significantly protected the renal tubule cell viability and prevented inflammatory signals against H2O2 challenge. LIPUS could inhibit the apoptosis-related molecular signals and increase the protein levels of endogenous antioxidant enzymes, α-Klotho, and Sirt1 in renal tubule cells after H/R challenge. LIPUS alleviated the increases in the serum levels of blood urea nitrogen, creatinine, and cystatin C, renal pathological changes and apoptosis-related molecular signals, and impaired antioxidant enzymes in AKI mice. The IRI-induced inflammatory responses in the kidneys and spleens could be reversed by LIPUS. These findings suggest that LIPUS treatment displays the benefits for renal protection in IRI-induced AKI mice.


Subject(s)
Acute Kidney Injury/prevention & control , Reperfusion Injury/prevention & control , Ultrasonic Therapy , Animals , Apoptosis/radiation effects , Cell Line , Inflammation/therapy , Rats
11.
Sci Rep ; 10(1): 9928, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32555254

ABSTRACT

Silicon dioxide nanoparticles (SiO2NPs) are widely applied in industry, chemical, and cosmetics. SiO2NPs is known to induce pulmonary toxicity. In this study, we investigated the molecular mechanisms of SiO2NPs on pulmonary toxicity using a lung alveolar epithelial cell (L2) model. SiO2NPs, which primary particle size was 12 nm, caused the accumulation of intracellular Si, the decrease in cell viability, and the decrease in mRNAs expression of surfactant, including surfactant protein (SP)-A, SP-B, SP-C, and SP-D. SiO2NPs induced the L2 cell apoptosis. The increases in annexin V fluorescence, caspase-3 activity, and protein expression of cleaved-poly (ADP-ribose) polymerase (PARP), cleaved-caspase-9, and cleaved-caspase-7 were observed. The SiO2NPs induced caspase-3 activity was reversed by pretreatment of caspase-3 inhibitor Z-DEVD-FMK. SiO2NPs exposure increased reactive oxygen species (ROS) production, decreased mitochondrial transmembrane potential, and decreased protein and mRNA expression of Bcl-2 in L2 cells. SiO2NPs increased protein expression of cytosolic cytochrome c and Bax, and mRNAs expression of Bid, Bak, and Bax. SiO2NPs could induce the endoplasmic reticulum (ER) stress-related signals, including the increase in CHOP, XBP-1, and phospho-eIF2α protein expressions, and the decrease in pro-caspase-12 protein expression. SiO2NPs increased phosphoinositide 3-kinase (PI3K) activity and AKT phosphorylation. Both ROS inhibitor N-acetyl-l-cysteine (NAC) and PI3K inhibitor LY294002 reversed SiO2NPs-induced signals described above. However, the LY294002 could not inhibit SiO2NPs-induced ROS generation. These findings demonstrated first time that SiO2NPs induced L2 cell apoptosis through ROS-regulated PI3K/AKT signaling and its downstream mitochondria- and ER stress-dependent signaling pathways.


Subject(s)
Alveolar Epithelial Cells/pathology , Apoptosis , Endoplasmic Reticulum Stress/drug effects , Mitochondria/pathology , Nanoparticles/administration & dosage , Oxidative Stress , Silicon Dioxide/pharmacology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Cell Survival , Cells, Cultured , Gene Expression Regulation , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Nanoparticles/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction
12.
EJNMMI Res ; 10(1): 13, 2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32096011

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) are involved in drug resistance, metastasis, and relapse of cancers, which can significantly affect tumor therapy. Hence, to develop specifically therapeutic target probe at CSCs for improvement of survival and quality of life of cancer patients is urgently needed. The CD166 protein has been suggested to be involved in colorectal cancer (CRC) tumorigenesis and to be considered a marker for colorectal CSCs (CRCSCs) detection. In this study, therefore, we attend to apply a nuclear imaging agent probe, Glycine18-Cystine-linked CD166-targeted peptides (CD166tp-G18C), to detect the changes of CD166 level in a CRC xenograft mouse model. RESULTS: We isolated the CD166-positive cells from the HCT15 CRC cell line (CD166+HCT15) and evaluated their morphology and ability of clone formation, migration, protein expression, and drug resistance. The CD166-positive HCT15 cells display the CSCs characteristics. We discovered and designed a CD166-targeted peptide (CD166tp-G18C) as a targeted probe of CRC stem-like cell for cell binding assay. The CD166tp-G18C confirmed the CD166 protein targeting ability in CD166+HCT15 cells. The diethylenetriaminopentaacetic acid (DTPA)-conjugated CD166tp-G18C further was labeled with indium-111 (111In-DTPA-CD166tp-G18C) as nuclear imaging agent for imaging and bio-distribution analysis in vivo. Finally, we observed that the 111In-DTPA-CD166tp-G18C was significantly enhanced in tumor tissues of CD166+HCT15 xenograft mice as compared to the non-CD166tp-G18C control. CONCLUSIONS: Our results indicated that the indium-111-labeled CD166tp-G18C may be served as a powerful tool for colorectal CSCs nuclear imaging in the CRC patients.

13.
Toxicol In Vitro ; 63: 104739, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31756540

ABSTRACT

Human exposure to silica nanoparticles (SiNPs) has been widely applied as vehicles for drug delivery and cellular manipulations in nanoneuromedicine. SiNPs may cause adverse effects in the brain, but potential mechanisms underlying SiNPs-induced neurotoxicity are remained unclear. Here, we examined cytotoxic effects and the cellular mechanisms of SiNPs-induced neuronal cell death. In this study, the results showed that SiNPs significantly decreased cell viability and induced apoptosis in Neuro-2a cells as evidenced by the increase caspase-3 activity and the activation of caspase cascades and poly (ADP-ribose) polymerase (PARP). In addition, endoplasmic reticulum (ER) stress was triggered as indicated by several key molecules including glucose-regulated protein (GRP)78 and 94, C/EBP homologous protein (CHOP), activation transcription factor (ATF)-4, and caspase-12. Pretreatment of Neuro-2a cells with specific pharmacological inhibitor of ER stress (4-phenylbutyric acid (4-PBA)) effectively alleviated the SiNPs-induced ER stress and apoptotic related signals. Furthermore, 2',7'-Dichlorofluorescein fluorescence as an indicator of reactive oxygen species (ROS) formation after exposure of Neuro-2a cells to SiNPs significantly increased ROS levels. Antioxidant N-acetylcyseine (NAC) effectively reversed SiNPs-induced cellular responses. Taken together, these results suggest that SiNPs exposure exerts its neurotoxicity in cultured neuronal cells by inducing apoptosis via a ROS generation-activated downstream ER stress signaling pathway.


Subject(s)
Nanoparticles/toxicity , Neurons/drug effects , Silicon Dioxide/toxicity , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , Neurons/metabolism , Reactive Oxygen Species/metabolism
14.
Int J Mol Sci ; 20(20)2019 Oct 18.
Article in English | MEDLINE | ID: mdl-31635269

ABSTRACT

The incidence of stroke recurrence is still higher despite the advanced progression of therapeutic treatment and medical technology. Low intensity pulsed ultrasound (LIPUS) has been demonstrated to possess therapeutic effects on neuronal diseases and stroke via brain-derived neurotrophic factor (BDNF) induction. In this study, we hypothesized that LIPUS treatment possessed therapeutic benefits for the improvement of stroke recurrence. Adult male C57BL/6J mice were subjected to a middle cerebral artery occlusion (MCAO) surgery and then followed to secondary MCAO surgery as a stroke recurrence occurred after nine days from the first MCAO. LIPUS was administered continuously for nine days before secondary MCAO. LIPUS treatment not only decreased the mortality but also significantly moderated neuronal function injury including neurological score, motor activity, and brain pathological score in the recurrent stroke mice. Furthermore, the administration of LIPUS attenuated the apoptotic neuronal cells and increased Bax/Bcl-2 protein expression ratio and accelerated the expression of BDNF in the brain of the recurrent stroke mice. Taken together, these results demonstrate for the first time that LIPUS treatment arouses the expression of BDNF and possesses a therapeutic benefit for the improvement of stroke recurrence in a mouse model. The neuroprotective potential of LIPUS may provide a useful strategy for the prevention of a recurrent stroke.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Reperfusion Injury/prevention & control , Reperfusion Injury/therapy , Stroke/etiology , Stroke/prevention & control , Ultrasonic Therapy , Ultrasonic Waves , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Biomarkers , Disease Models, Animal , Immunohistochemistry , Male , Mice , Motor Activity , Reperfusion Injury/etiology , Reperfusion Injury/mortality , Stroke/diagnosis , Stroke/therapy
15.
Vascul Pharmacol ; 121: 106578, 2019 10.
Article in English | MEDLINE | ID: mdl-31279923

ABSTRACT

Benzo[a]pyrene (BaP) is a polycyclic aromatic hydrocarbon found in tobacco smoke and air pollution products. BaP exposure has been recently suggested to be a risk factor for hypertension in coke oven workers. The mechanisms of BaP on vascular smooth muscle function remain unclear. Here, we examined the influence and possible mechanism of BaP on vasoconstriction in rat thoracic aortas ex vivo and in vivo. In vivo exposure of rats to BaP (20 mg/kg) for 8 weeks caused a significant enhancement in the systolic blood pressure and enhanced aortic hyperreactivity to α1-adrenoceptor selective agonist phenylephrine in aortas. BaP (1 and 10 µM) treatment for 18 h induced an enhancement of phenylephrine-induced vasoconstriction in the organ cultures of aortas. Aryl hydrocarbon receptor antagonist α-naphthoflavone, protein kinase C (PKC) inhibitor chelerythrine, mitogen-activated protein kinases (MAPK) inhibitor PD98059, myosin light chain kinase (MLCK) inhibitor ML-9, and Rho-kinase inhibitor Y-27632 significantly suppressed BaP-enhanced vasoconstriction. BaP time-dependently triggered reactive oxygen species (ROS) production in primary vascular smooth muscle cells. Both antioxidant N-acetylcysteine and NAD(P)H oxidase inhibitor diphenyleneiodonium significantly inhibited BaP-triggered ROS production and vasoconstriction. These results suggest that BaP enhances aortic vasoconstriction via the activation of ROS and muscular signaling molecules PKC, MAPK, MLCK, and Rho-kinase.


Subject(s)
Benzo(a)pyrene/toxicity , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Vasoconstriction/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Cells, Cultured , In Vitro Techniques , Male , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myosin-Light-Chain Kinase/metabolism , Protein Kinase C/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction , rho-Associated Kinases/metabolism
16.
Cells ; 8(7)2019 06 30.
Article in English | MEDLINE | ID: mdl-31262060

ABSTRACT

Diabetes-associated advanced glycation end-products (AGEs) can increase extracellular matrix (ECM) expression and induce renal fibrosis. Calbindin-D28k, which plays a role in calcium reabsorption in renal distal convoluted tubules, is increased in a diabetic kidney. The role of calbindin-D28k in diabetic nephropathy still remains unclear. Here, calbindin-D28k protein expression was unexpectedly induced in the renal tubules of db/db diabetic mice. AGEs induced the calbindin-D28k expression in human renal proximal tubule cells (HK2), but not in mesangial cells. AGEs induced the expression of fibrotic molecules, ECM proteins, epithelial-mesenchymal transition (EMT) markers, and endoplasmic reticulum (ER) stress-related molecules in HK2 cells, which could be inhibited by a receptor for AGE (RAGE) neutralizing antibody. Calbindin-D28k knockdown by siRNA transfection reduced the cell viability and obviously enhanced the protein expressions of fibrotic factors, EMT markers, and ER stress-related molecules in AGEs-treated HK2 cells. Chemical chaperone 4-Phenylbutyric acid counteracted the AGEs-induced ER stress and ECM and EMT markers expressions. Calbindin-D28k siRNA in vivo delivery could enhance renal fibrosis in db/db diabetic mice. These findings suggest that inducible calbindin-D28k protects against AGEs/RAGE axis-induced ER stress-activated ECM induction and cell injury in renal proximal tubule cells.


Subject(s)
Calbindin 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/pathology , Glycation End Products, Advanced/metabolism , Kidney Tubules, Distal/pathology , Animals , Calbindin 1/genetics , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Fibrosis , Gene Knockdown Techniques , Humans , Kidney Tubules, Distal/drug effects , Male , Mesangial Cells/metabolism , Mice , Phenylbutyrates/pharmacology , RNA, Small Interfering/metabolism , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/metabolism
17.
Phytomedicine ; 59: 152917, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30978648

ABSTRACT

BACKGROUND: The pathology change of renal tubulointerstitial fibrosis is a critical feature of chronic kidney disease (CKD), regardless of the primary insults. The infiltration of inflammatory cells and the consecutive secretion of profibrotic factors are frequently and conspicuously observed during the development of renal fibrosis. Icariin, an active polyphenol of the Epimedium genus, has been found to alleviate the symptoms of chronic diseases like diabetes, neurodegeneration, and heart and renal diseases. The effect and mechanism of icariin on the prevention of CKD-associated renal fibrosis still needed clarification. PURPOSE: The aims of this study were to investigate whether icariin treatment improves the development of CKD-associated renal fibrosis and its possible mechanism. METHODS: An experimental unilateral ureteral obstruction (UUO)-induced chronic renal fibrosis mouse model was used. Mice were orally administered with icariin (20 mg/kg/day) for 3 consecutive days before and 14 consecutive days after UUO surgery. RESULTS: The pathological changes, collagen deposition, and protein expressions of profibrotic factors (transforming growth factor-ß and connective tissue growth factor) and fibrotic markers (α-smooth muscle actin and fibronectin), which were significantly elevated in the kidneys of UUO mice, could be significantly reversed by icariin treatment. Icariin treatment also significantly inhibited the increased Smad2/3 and decreased E-cadherin protein expressions in the kidneys of UUO mice. Icariin treatment prominently mitigated the protein expression of proinflammatory factors like nuclear factor-κB, cyclooxygenase-2, interleukin 1-ß and prooxidative enzyme (NADPH oxidase-4), and it increased the protein expression of antioxidative enzymes (superoxide dismutase and catalase). CONCLUSION: Icariin treatment protects against CKD-associated renal fibrosis via its antifibrotic and anti-inflammatory properties. Icariin may serve as a therapeutic agent in the prevention of CKD-associated renal fibrosis.


Subject(s)
Flavonoids/therapeutic use , Kidney Diseases/drug therapy , Kidney/drug effects , Ureteral Obstruction/etiology , Animals , Anti-Inflammatory Agents/pharmacology , Collagen/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis/etiology , Fibrosis/prevention & control , Male , Mice , Transforming Growth Factor beta/metabolism , Ureteral Obstruction/complications
18.
J Orthop Res ; 37(4): 942-947, 2019 04.
Article in English | MEDLINE | ID: mdl-30835896

ABSTRACT

Expression of CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP) is induced during endoplasmic reticulum (ER) stress, which is related to apoptosis in several cell types. CHOP null mice have been exhibited to decrease bone formation. However, a study of transgenic mice overexpressing CHOP in the bone microenvironment showed that CHOP overexpression impairs the osteoblastic function leading to osteopenia. The regulatory role of CHOP in bone formation is controversial and still remains to be clarified. Here, we investigated the alterations in bone microstructure of CHOP knockout (Chop-/- ) mice and tested the gender difference of CHOP deficiency in susceptibility to osteopenia. Adult female and male mice (WT) and Chop-/- mice were used. The microcomputed tomography (µCT) analysis in trabecular bone and cortical bone of tibia was determined. Trabecular bone volume fraction (BV/TV), trabecular number, and bone mineral density (BMD) in tibia are markedly decreased in both male and female Chop-/- mice compared to the control WT mice. Unexpectedly, the BMD and BV/TV in trabecular bone of tibia in female Chop-/- mice were significantly lower than in male Chop-/- mice. The similar results could also be observed in the cortical bone of tibia in Chop-/- mice. This gender difference was also observed in the decreased capacity of osteoblast differentiation of bone marrow cells isolated from Chop-/- mice. These results indicated that ER stress-related CHOP signaling might play an important role in the bone formation in a mouse model, especially in females. There is the gender difference of CHOP deficiency in susceptibility to osteopenia. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.


Subject(s)
Bone Diseases, Metabolic/etiology , Sex Characteristics , Transcription Factor CHOP/physiology , Animals , Bone Density , Bone Diseases, Metabolic/diagnostic imaging , Female , Male , Mice, Knockout , X-Ray Microtomography
19.
J Transl Med ; 16(1): 168, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29921305

ABSTRACT

BACKGROUND: The human epidermal growth factor receptor 2 (HER2) involved proliferation, angiogenesis, and reduced apoptosis in gastric cancer (GC), which is a common target for tumor therapy. HER2 is usually overexpressed in more than 15% GC patients, developing a reliable diagnostic tool for tumor HER2 detection is important. In this study, we attend to use polyethylene glycol (PEG) linked anti-HER2/neu peptide (AHNP-PEG) as a nuclear imaging agent probe for HER2 detection in GC xenograft animal model. METHODS: The HER2 expression of human sera and tissues were detected in GC patients and normal subjects. GC cell lines NCI-N87 (high HER2 levels) and MKN45 (low HER2 levels) were treated with AHNP-PEG to assess the cell viability and HER2 binding ability. The NCI-N87 was treated with AHNP-PEG to observe the level and phosphorylation of HER2. The MKN45 and NCI-N87-induced xenograft mice were intravenous injection with fluorescence labeled AHNP-PEG for detecting in vivo fluorescence imaging properties and biodistribution. The AHNP-PEG was conjugated with diethylenetriaminopentaacetic acid (DTPA) for indium-111 labeling (111In-DTPA-AHNP-PEG). The stability of was assessed in vitro. The imaging properties and biodistribution of 111In-DTPA-AHNP-PEG were observed in NCI-N87-induced xenograft mice. RESULTS: The serum HER2 (sHER2) levels in GC patients were significantly higher than the normal subjects. The sHER2 levels were correlated with the tumor HER2 levels in different stages of GC patients. The AHNP-PEG inhibited the cell growth and down-regulated HER2 phosphorylation in HER2-overexpressed human GC cells (NCI-N87) via specific HER2 interaction of cell surface. In addition, the GC tumor tissues from HER2-postive xenograft mice presented higher HER2 fluorescence imaging as compared to HER2-negative group. The HER2 levels in the tumor tissues were also higher than other organs in NCI-N87-induced xenograft mice. Finally, we further observed that the 111In-DTPA-AHNP-PEG was significantly enhanced in tumor tissues of NCI-N87-induced xenograft mice compared to control. CONCLUSIONS: These findings suggest that the sHER2 measurement may be as a potential tool for detecting HER2 expressions in GC patients. The radioisotope-labeled AHNP-PEG may be useful to apply in GC patients for HER2 nuclear medicine imaging.


Subject(s)
Molecular Probes/chemistry , Peptides/chemistry , Polyethylene Glycols/chemistry , Receptor, ErbB-2/metabolism , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/diagnosis , Animals , Cell Line, Tumor , Cell Survival , Female , Humans , Indium Radioisotopes/chemistry , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Organometallic Compounds/chemistry , Phosphorylation , Receptor, ErbB-2/blood , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Xenograft Model Antitumor Assays
20.
Toxicol Sci ; 164(1): 363-374, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29669060

ABSTRACT

Plasticizer di-(2-ethylhexyl)phthalate (DEHP) is known as an endocrine disruptor and a peroxisome proliferator. A currently epidemiological study has suggested that daily high DEHP intake from phthalate-tainted foods in children may be a risk factor for renal dysfunction. DEHP can leach from medical devices such as blood storage bags and the tubing. Long-term exposure to DEHP is associated with nephropathy and exacerbates chronic kidney diseases (CKDs) progression. However, the detailed effects and molecular mechanisms remain unclear. Here, we hypothesized that DEHP and its major metabolite mono-(2-ethylhexyl)phthalate (MEHP) incited epithelial-to-mesenchymal transition (EMT) and lead to aggravate renal fibrosis progression. Treatment with low-cytotoxic concentration DEHP, but not MEHP, for 72 h obviously induced the morphological and phenotypic changes and EMT markers induction in normal rat renal tubular epithelial cells (NRK-52E). AKT inhibitor MK-2206 inhibited DEHP-induced EMT features and signals of AKT phosphorylation and downstream NF-κB and GSK3ß. DEHP did not affect the expression of transforming growth factor-ß1 mRNA. DEHP down-regulated the peroxisome proliferator-activated receptor (PPAR)α and PPARγ protein expressions. PPARγ agonist pioglitazone partially and significantly inhibited DEHP-induced EMT induction. In vivo DEHP exposure for 6 weeks enhanced the renal dysfunction and renal fibrosis and mortality rate, but decreased the PPARα and PPARγ protein expressions, in a folic acid-induced kidney fibrosis mouse model. Taken together, these results demonstrate for the first time that DEHP arouses EMT induction and renal fibrosis progression in renal tubular cells and is associated with PPARs downregulation. DEHP exposure potentially exacerbated renal fibrosis/nephropathy in a kidney disease condition.


Subject(s)
Diethylhexyl Phthalate/toxicity , Endocrine Disruptors/toxicity , Epithelial-Mesenchymal Transition/drug effects , Kidney/drug effects , Kidney/pathology , Plasticizers/toxicity , Animals , Cell Line , Cell Survival/drug effects , Fibrosis , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Mice, Inbred C57BL , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...