Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
J Nanobiotechnology ; 22(1): 56, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38336783

ABSTRACT

Diabetic retinopathy (DR) is a vision-threatening diabetic complication that is characterized by microvasculature impairment and immune dysfunction. The present study demonstrated that M2 microglia intensively participated in retinal microangiopathy in human diabetic proliferative membranes, mice retinas, retinas of mice with oxygen-induced retinopathy (OIR) mice, and retinas of streptozotocin-induced DR mice. Further in vivo and in vitro experiments showed that exosomes derived from M2 polarized microglia (M2-exo) could reduce pericyte apoptosis and promote endothelial cell proliferation, thereby promoting vascular remodeling and reducing vascular leakage from the diabetic retina. These effects were further enhanced by M2-exo that facilitated M2 polarization of retinal microglia. Collectively, the study demonstrated the capability of M2-exo to induce retinal microvascular remodeling, which may provide a new therapeutic strategy for the treatment of DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Exosomes , Mice , Animals , Humans , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/etiology , Vascular Remodeling , Microglia , Retina
2.
Redox Biol ; 69: 103030, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181705

ABSTRACT

Ferroptosis is a type of programmed cell death resulting from iron overload-dependent lipid peroxidation, and could be promoted by activating transcription factor 3 (ATF3). SIRT1 is an enzyme accounting for removing acetylated lysine residues from target proteins by consuming NAD+, but its role remains elusive in ferroptosis and activating ATF3. In this study, we found SIRT1 was activated during the process of RSL3-induced glioma cell ferroptosis. Moreover, the glioma cell death was aggravated by SIRT1 activator SRT2183, but suppressed by SIRT inhibitor EX527 or when SIRT1 was silenced with siRNA. These indicated SIRT1 sensitized glioma cells to ferroptosis. Furthermore, we found SIRT1 promoted RSL3-induced expressional upregulation and nuclear translocation of ATF3. Silence of ATF3 with siRNA attenuated RSL3-induced increases of ferrous iron and lipid peroxidation, downregulation of SLC7A11 and GPX4 and depletion of cysteine and GSH. Thus, SIRT1 promoted glioma cell ferroptosis by inducting ATF3 activation. Mechanistically, ATF3 activation was reinforced when RSL3-induced decline of NAD+ was aggravated by FK866 that could inhibit NAD + synthesis via salvage pathway, but suppressed when intracellular NAD+ was maintained at higher level by supplement of exogenous NAD+. Notably, the NAD + decline caused by RSL3 was enhanced when SIRT1 was further activated by SRT2183, but attenuated when SIRT1 activation was inhibited by EX527. These indicated SIRT1 promoted ATF3 activation via consumption of NAD+. Finally, we found RSL3 activated SIRT1 by inducing reactive oxygen species-dependent upregulation of AROS. Together, our study revealed SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via activation of ATF3-dependent inhibition of SLC7A11 and GPX4.


Subject(s)
Ferroptosis , Glioma , Humans , NAD , Activating Transcription Factor 3/genetics , Cell Line, Tumor , Sirtuin 1/genetics , Glioma/genetics , Glioma/metabolism , RNA, Small Interfering
3.
Neurosurg Rev ; 47(1): 35, 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38183517

ABSTRACT

Clear cell meningiomas are a rare histological subtype of World Health Organization (WHO) grade II meningioma. Despite its relatively low frequency, clear cell meningioma has attracted considerable attention because of its unique pathological characteristics, clinical behavior, and challenging management considerations. The purpose of our systematic review is to provide clinicians with a better understanding of this rare disease. PubMed was searched for articles in the English language published from 1988 to 2023 June. The keywords were as follows: "clear cell meningioma," "clear cell" and "meningioma." We analyzed clinical manifestations, radiological manifestations, pathological features, comprehensive treatment strategies, and prognosis to determine the factors influencing recurrence-free survival (RFS). Recurrence-free survival curves of related factors were calculated by the Kaplan‒Meier method. The log-rank test and Cox univariate analysis were adopted to assess the intergroup differences and seek significant factors influencing prognosis and recurrence. Fifty-seven papers met the eligibility criteria, including 207 cases of clear cell meningioma (CCM), which were confirmed by postoperative pathology. The fifty-seven articles involved 84 (40.6%) males and 123 (59.4%) females. The average age at diagnosis was 27.9 years (range, 14 months to 84 years). Among the symptoms observed, headache, neurologic deficit, and hearing loss were the most commonly reported clinical manifestations. Most tumors (47.8%) were located in the skull base region. Most tumors showed significant enhancement, and homogeneous enhancement was more common. A total of 152 (74.1%) patients underwent gross total resection (GTR), and 53 (25.9%) patients underwent subtotal resection (STR). During the follow-up, the tumor recurred in 80 (39.4%) patients. The log-rank test and the Cox univariate analysis revealed that tumor resection range (GTR vs. STR) and adjuvant treatment (YES vs. NO) were significant predictors of recurrence-free survival (RFS). Clear cell meningioma is a rare type of meningioma with challenging diagnosis and therapy. The prognosis of this disease is different from that of regular meningiomas. Recurrence remains a possibility even after total tumor resection. We found that the surgical resection range and adjuvant treatment affected the recurrence period. This finding provides significant guidance for the treatment of clear cell meningioma.


Subject(s)
Meningeal Neoplasms , Meningioma , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Young Adult , Central Nervous System , Headache , Meningeal Neoplasms/surgery , Meningioma/surgery
4.
Environ Sci Pollut Res Int ; 30(56): 119095-119116, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37919494

ABSTRACT

The Green Financial Reform and Innovation Pilot Areas (GFRIPA) policy is a key institutional arrangement that enables China's green finance to advance from theory to practice. Few studies have quantitatively evaluated the policy's environmental performance. This study uses a generalized synthetic control method (GSCM) alongside panel data from Chinese prefecture-level cities since 2007 to assess the effects of the GFRIPA policy on energy consumption and pollution emissions and to pinpoint the underlying mechanisms. Results show that establishing the GFRIPA significantly reduces energy consumption and pollution emissions, and that the effect emerges immediately in the policy's issuance year. Possible mechanisms consist of the increase in urban green innovation, the ease of financing constraints, the optimization of industrial structure, and the enhancement of environmental governance. Heterogeneity analyses reveal that policy effects are more profound in cities with a higher degree of marketization and a higher level of education. The findings provide valuable insights into consistently promoting the GFRIPA policy to meet environmental goals for energy conservation and pollution reduction and ultimately advance green economies in developing nations.


Subject(s)
Conservation of Natural Resources , Fiscal Policy , China , Cities , Economic Development , Environmental Policy , Policy
7.
ACS Appl Mater Interfaces ; 15(25): 29789-29800, 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37328300

ABSTRACT

Antioxidants are crucial for human health, and the detection of antioxidants can provide valuable information for disease diagnosis and health management. In this work, we report a plasmonic sensing approach for the determination of antioxidants based on their antietching capacity toward plasmonic nanoparticles. The Ag shell of core-shell Au@Ag nanostars can be etched by chloroauric acid (HAuCl4), whereas antioxidants can interact with HAuCl4, which prevents the surface etching of Au@Ag nanostars. We modulate the thickness of the Ag shell and morphology of the nanostructures, showing that the core-shell nanostars with the smallest thickness of Ag shell have the best etching sensitivity. Owing to the extraordinary surface plasmon resonance (SPR) property of Au@Ag nanostars, the antietching effect of antioxidants can induce a significant change in both the SPR spectrum and the color of solution, facilitating both the quantitative detection and naked-eye readout. This antietching strategy enables the determination of antioxidants such as cystine and gallic acid with a linear range of 0.1-10 µM. The core-shell Au@Ag nanostars are further immobilized in agarose gels to fabricate test strips, which can display different color changes in the presence of HAuCl4 from 0 to 1000 µM. The agarose-based test strip is also capable of detecting antioxidants in real samples, which allows naked-eye readout and quantitative detection by a smartphone.


Subject(s)
Antioxidants , Metal Nanoparticles , Humans , Point-of-Care Systems , Sepharose , Gold/chemistry , Metal Nanoparticles/chemistry , Silver/chemistry
8.
Acta Pharmacol Sin ; 44(10): 2125-2138, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37277492

ABSTRACT

Parthanatos is a type of programmed cell death dependent on hyper-activation of poly (ADP-ribose) polymerase 1 (PARP-1). SIRT1 is a highly conserved nuclear deacetylase and often acts as an inhibitor of parthanatos by deacetylation of PARP1. Our previous study showed that deoxypodophyllotoxin (DPT), a natural compound isolated from the traditional herb Anthriscus sylvestris, triggered glioma cell death via parthanatos. In this study, we investigated the role of SIRT1 in DPT-induced human glioma cell parthanatos. We showed that DPT (450 nmol/L) activated both PARP1 and SIRT1, and induced parthanatos in U87 and U251 glioma cells. Activation of SIRT1 with SRT2183 (10 µmol/L) enhanced, while inhibition of SIRT1 with EX527 (200 µmol/L) or knockdown of SIRT1 attenuated DPT-induced PARP1 activation and glioma cell death. We demonstrated that DPT (450 nmol/L) significantly decreased intracellular NAD+ levels in U87 and U251 cells. Further decrease of NAD+ levels with FK866 (100 µmol/L) aggravated, but supplement of NAD+ (0.5, 2 mmol/L) attenuated DPT-induced PARP1 activation. We found that NAD+ depletion enhanced PARP1 activation via two ways: one was aggravating ROS-dependent DNA DSBs by upregulation of NADPH oxidase 2 (NOX2); the other was reinforcing PARP1 acetylation via increase of N-acetyltransferase 10 (NAT10) expression. We found that SIRT1 activity was improved when being phosphorylated by JNK at Ser27, the activated SIRT1 in reverse aggravated JNK activation via upregulating ROS-related ASK1 signaling, thus forming a positive feedback between JNK and SIRT1. Taken together, SIRT1 activated by JNK contributed to DPT-induced human glioma cell parthanatos via initiation of NAD+ depletion-dependent upregulation of NOX2 and NAT10.


Subject(s)
Glioma , Parthanatos , Sirtuin 1 , Humans , Glioma/drug therapy , N-Terminal Acetyltransferases/genetics , N-Terminal Acetyltransferases/metabolism , NAD/metabolism , NADPH Oxidase 2/metabolism , Parthanatos/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Up-Regulation
9.
Acta Pharmacol Sin ; 44(9): 1906-1919, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37186123

ABSTRACT

Parthanatos is a type of programmed cell death initiated by over-activated poly (ADP-ribose) polymerase 1 (PARP1). Nuclear translocation of apoptosis inducing factor (AIF) is a prominent feature of parthanatos. But it remains unclear how activated nuclear PARP1 induces mitochondrial AIF translocation into nuclei. Evidence has shown that deoxypodophyllotoxin (DPT) induces parthanatos in glioma cells via induction of excessive ROS. In this study we explored the downstream signal of activated PARP1 to induce nuclear translocation of AIF in DPT-triggered glioma cell parthanatos. We showed that treatment with DPT (450 nM) induced PARP1 over-activation and Tax1 binding protein 1 (TAX1BP1) distribution to mitochondria in human U87, U251 and U118 glioma cells. PARP1 activation promoted TAX1BP1 distribution to mitochondria by depleting nicotinamide adenine dinucleotide (NAD+). Knockdown of TAX1BP1 with siRNA not only inhibited TAX1BP1 accumulation in mitochondria, but also alleviated nuclear translocation of AIF and glioma cell death. We demonstrated that TAX1BP1 enhanced the activity of respiratory chain complex I not only by upregulating the expression of ND1, ND2, NDUFS2 and NDUFS4, but also promoting their assemblies into complex I. The activated respiratory complex I generated more superoxide to cause mitochondrial depolarization and nuclear translocation of AIF, while the increased mitochondrial superoxide reversely reinforced PARP1 activation by inducing ROS-dependent DNA double strand breaks. In mice bearing human U87 tumor xenograft, administration of DPT (10 mg· kg-1 ·d-1, i.p., for 8 days) markedly inhibited the tumor growth accompanied by NAD+ depletion, TAX1BP1 distribution to mitochondria, AIF distribution to nuclei as well as DNA DSBs and PARP1 activation in tumor tissues. Taken together, these data suggest that TAX1BP1 acts as a downstream signal of activated PARP1 to trigger nuclear translocation of AIF by activation of mitochondrial respiratory chain complex I.


Subject(s)
Glioma , Parthanatos , Humans , Mice , Animals , Apoptosis Inducing Factor/genetics , Superoxides/metabolism , Reactive Oxygen Species/metabolism , NAD/metabolism , Electron Transport , Electron Transport Complex I , Glioma/metabolism , Neoplasm Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
10.
J Environ Manage ; 339: 117915, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37058925

ABSTRACT

Promoting green financial reform is an important measure to support environmentally-biased technological progress (EBTP) and achieve sustainable economic and social development. Although China launched a green finance reform and innovation pilot zone (GFRIPZ) policy in 2017, little is known about whether and how such a policy affects EBTP. Based on mathematical deduction, this paper studies the mechanism through which green financial reform influences EBTP. The analysis employs panel data of Chinese prefecture-level cities and a generalized synthetic control method to examine the policy effect of the establishment of GFRIPZ in EBTP. It is found that establishing GFRIPZ significantly promotes EBTP, and that the policy effect shows "ahead-of-policy" and dynamically increasing features. Potential mechanisms reside in the pilot policy's easing of financing constraints and upgrading of industrial structure. Further heterogeneity analyses reveal that great disparities exist in the policy effects of different pilot zones, with a steadily increasing policy effect in Zhejiang and Guangdong, a lagging policy effect in Jiangxi and Guizhou, and an inverse U-shaped policy effect in Xinjiang. Policy effects are much stronger in regions with a higher degree of marketization and a higher level of attention to education. Additional tests of economic performance indicate that the pilot policy, interweaved with its driving effect on EBTP, is conducive to promoting an energy-conservation and low-carbon-energy transition. The findings shed light on applying green financial reform to encourage environment-friendly technological research and development.


Subject(s)
Carbon , Industry , China , Cities , Policy , Economic Development
11.
Mol Med Rep ; 27(3)2023 Mar.
Article in English | MEDLINE | ID: mdl-36799163

ABSTRACT

Maltol, a chemical isolated from ginseng root, has shown treatment effects on several pathological processes including osteoarthritis, diabetic peripheral neuropathy and liver fibrosis. Nevertheless, its effect on ischemia­induced neuron death remains elusive. In the present study, the treatment effect of maltol on ischemia­induced neuron damage was investigated by using oxygen and glucose deprivation (OGD) model in SH­SY5Y cells. In vitro studies revealed that maltol protected SH­SY5Y cells against OGD­induced chromatinolysis by inhibiting two reactive oxygen species (ROS)­regulated pathways. One was DNA double­strand breaks and the other was nuclear translocation of apoptosis inducing factor. Mechanistically, maltol not only inhibited OGD­induced depletion of glutathione and cysteine by maintaining cystine/glutamate antiporter (xCT) level, but also abrogated OGD­induced catalase downregulation. Meanwhile, maltol also alleviated OGD­induced inactivation of mTOR by attenuating OGD­induced depletion of adenosine triphosphate and pyruvate and downregulation of pyruvate kinase M2, indicating that maltol inhibited the glycolysis dysfunction caused by OGD. Considering that activated mammalian target of the rapamycin (mTOR) could lead to enhanced xCT expression and decreased catalase degradation by autophagy, these findings indicated that maltol attenuated OGD­induced ROS via inhibition of mTOR inactivation by maintaining pyruvate level. Taken together, it was demonstrated that maltol prevented OGD­induced chromatinolysis in SH­SY5Y cells via inhibiting pyruvate depletion.


Subject(s)
Neuroblastoma , Oxygen , Humans , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Pyruvic Acid , Glucose/metabolism , Catalase , TOR Serine-Threonine Kinases/metabolism
12.
Int Immunopharmacol ; 114: 109481, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36470119

ABSTRACT

Effective treatment of liver fibrosis remains a challenging medical problem. Taraxasterol (TAR) has anti-inflammatory, anti-tumor and hepatoprotective effects. Studies have shown that TAR has good biological activity against liver injury induced by various factors. However, the anti-fibrotic effect of TAR and its mechanism are never clarified. The purpose of this study was to investigate the effects of TAR in liver fibrosis and to reveal its possible mechanism by RNA sequencing. Our results suggested that TAR attenuated CCl4-induced hepatocyte necrosis, inflammatory infiltration and ECM deposition. TAR inhibited the levels of ALT, AST, ALP, γ-GT, LN, HA, PC III and IV-C in serum and TNF-α, IL-6, IL-1ß and MDA in liver. In addition, TAR increased the activities of SOD and GSH-Px in liver. RNA sequencing analysis of liver tissues revealed that CCl4 and TAR significantly altered 4,155 genes and 2,675 genes, respectively. TAR reversed changes in ECM-related genes. More specifically, TAR mediated the expression of genes related to the activation of the Hippo pathway, while inhibiting the expression of genes related to the activation of HIF-1α, TGF-ß/Smad, and Wnt pathways. In the validation experiments, the qRT-PCR results showed that the expression levels of Yap1, Tead3, Hif1α, Vegfa, Tgfß1, Want3a, and Ctnnb1 mRNA were consistent with the RNA sequencing results. The Western blot results showed that TAR inhibited the levels of TGF-ß1 and p-Smad2. In addition, the results in vitro were consistent with those in vivo. Therefore, we concluded that TAR improved CCl4-induced liver fibrosis by regulating Hippo, HIF-1α, TGF-ß/Smad and Wnt pathways.


Subject(s)
Liver Cirrhosis , Liver , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver/pathology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta/metabolism , Sequence Analysis, RNA , Carbon Tetrachloride/adverse effects
13.
CNS Neurosci Ther ; 28(12): 2032-2043, 2022 12.
Article in English | MEDLINE | ID: mdl-35909335

ABSTRACT

AIMS: Epileptic seizures or status epilepticus (SE) can cause hippocampal neuronal death, which has detrimental effects. Parthanatos, a new form of programmed cell death, is characterized by hyperactivation of poly (ADP-ribose) polymerase-1 (PARP-1), excessive synthesis of poly ADP-ribose polymer, mitochondrial depolarization, and nuclear translocation of apoptosis-inducing factor, observed in various neurodegenerative disorders but rarely reported in epilepsy. We aimed to investigate whether parthanatos participates in the mechanism of seizure-induced hippocampal neuronal death. METHODS: Glutamate-mediated excitotoxicity cell model was used to study the mechanism of seizure-induced cell injury. Injection of kainic acid into the amygdala was used to establish the epileptic rat model. Corresponding biochemical tests were carried out on hippocampal tissues and HT22 cells following indicated treatments. RESULTS: In vitro, glutamate time-dependently induced HT22 cell death, accompanied by parthanatos-related biochemical events. Pretreatment with PJ34 (PARP-1 inhibitor) or small interfering RNA-mediated PARP-1 knockdown effectively protected HT22 cells against glutamate-induced toxic effects and attenuated parthanatos-related biochemical events. Application of the antioxidant N-acetylcysteine (NAC) rescued HT22 cell death and reversed parthanatos-related biochemical events. In vivo, PJ34 and NAC afforded protection against SE-induced hippocampal neuronal damage and inhibited parthanatos-related biochemical events. CONCLUSION: Parthanatos participates in glutamate-induced HT22 cell injury and hippocampal neuronal damage in rats following epileptic seizures. ROS might be the initiating factor during parthanatos.


Subject(s)
Parthanatos , Status Epilepticus , Rats , Animals , Kainic Acid , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Glutamic Acid , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/pharmacology , Cell Death , Hippocampus/metabolism , Acetylcysteine/pharmacology
14.
Dis Markers ; 2022: 5614939, 2022.
Article in English | MEDLINE | ID: mdl-35789603

ABSTRACT

To study the effects of ubenimex (UBE) combined with pemetrexed (PEM) on lung adenocarcinoma cell behavior and its molecular mechanism, the tissue samples from lung adenocarcinoma patients who received PEM chemotherapy, those with PEM combined with UBE chemotherapy, and healthy volunteers were retrieved and analyzed. The expression levels of the suppressor of cytokine signaling 1 (SOCS1) in the human lung adenocarcinoma cancer cell lines A549 and PC-9 and tissues were detected by qRT-PCR. MTT assay was performed for cell proliferation. Cell apoptosis was detected by flow cytometry. Cell invasion ability was assessed using the Transwell assay. The expression levels of the JAK2/STAT3 signaling pathway proteins and apoptosis-related proteins were detected by Western blot. The antitumor effect of PEM combined with UBE was tested in nude mice using the tumor formation assay. Our results showed that UBE treatment, alone or combined with PEM, inhibited lung adenocarcinoma cell migration, invasion, and proliferation; promoted apoptosis; significantly increased the G0/G1-phase cell ratio; reduced the S-phase cell ratio; and inhibited the in vivo growth of tumor cells. UBE alone or in combination with PEM also inhibited the JAK2/STAT3 signaling pathway in lung adenocarcinoma cells. In addition, UBE combined with PEM therapy was associated with increased SOCS1 expression in patients' serum and knocking down SOCS1 reversed the antitumor effects of UBE and PEM. Overall, combination therapy with UBE and PEM could inhibit the JAK2-STAT3 signaling pathway by upregulating SOCS1 expression to hinder the progression of lung adenocarcinoma cells.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma of Lung/drug therapy , Animals , Humans , Janus Kinase 2 , Leucine/analogs & derivatives , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Pemetrexed , STAT3 Transcription Factor , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/genetics
15.
Foodborne Pathog Dis ; 19(5): 349-358, 2022 05.
Article in English | MEDLINE | ID: mdl-35443788

ABSTRACT

Shigella flexneri, a common Gram-negative foodborne pathogen, is widely distributed in fresh-cut fruits and vegetables, unpasteurized milk, and food processing environments. The aims of this study were to evaluate the antibacterial effects of 405-nm light-emitting diode (LED) treatment on S. flexneri and to investigate the possible mechanism. The results showed that LED irradiation (360 min) reduced the number of S. flexneri in phosphate-buffered saline by 3.29 log colony-forming unit (CFU)/mL (initial bacterial count: 6.81 log CFU/mL). The cells in reconstituted infant formula, cells on fresh-cut carrot slices, and biofilm-associated cells on stainless steel surfaces were reduced by 1.83 log CFU/mL, 7.00 log CFU/cm2, and 4.35 log CFU/cm2 following LED treatment for 360, 120, and 120 min, respectively. LED treatment damaged both DNA and cell wall of S. flexneri and changed cell morphology and cell membrane permeability. In addition, LED treatment decreased total cell protein concentration of S. flexneri. These results indicated that 405-nm LED treatment effectively controlled S. flexneri contamination of foods and food contact surfaces and that the bacterial inactivation may be the result of damage to multiple cellular components. These findings highlight the potential of LED technology in controlling S. flexneri during food processing, storage, and preparation.


Subject(s)
Food Microbiology , Shigella flexneri , Colony Count, Microbial , Food Handling , Humans , Stainless Steel
16.
Biochem Biophys Res Commun ; 589: 1-8, 2022 01 22.
Article in English | MEDLINE | ID: mdl-34883284

ABSTRACT

BNIP3 is found to eliminate cancer cells via causing mitochondrial damage and endoplasmic reticulum stress, but it remains elusive of its role in regulating DNA double strand breaks (DSBs). In this study, we find that silibinin triggers DNA DSBs, ROS accumulation and expressional upregulation of BNIP3 in glioma cells. Mitigation of ROS with antioxidant GSH significantly inhibits silibinin-induced DNA DSBs and glioma cell death. Then, we find knockdown of BNIP3 with SiRNA obviously prevents silibinin-induced DNA DSBs and ROS accumulation. Mechanistically, BNIP3 knockdown not only reverses silibinin-triggered depletion of cysteine and GSH via maintaining xCT level, but also abrogates catalase decrease. Notably, silibinin-induced dephosphorylation of mTOR is also prevented when BNIP3 is knocked down. Given that activated mTOR could promote xCT expression and inhibit autophagic degradation of catalase, our data suggest that BNIP3 contributes to silibinin-induced DNA DSBs via improving intracellular ROS by inhibition of mTOR.


Subject(s)
DNA Breaks, Double-Stranded , Glioma/metabolism , Glioma/pathology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Silybin/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Transport System y+/metabolism , Catalase/metabolism , Cell Line, Tumor , Cysteine/metabolism , DNA Breaks, Double-Stranded/drug effects , Down-Regulation/drug effects , Glutathione/metabolism , Humans , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism
17.
Adv Healthc Mater ; 11(2): e2101926, 2022 01.
Article in English | MEDLINE | ID: mdl-34738742

ABSTRACT

Most of the antitumor chemotherapeutic drugs execute the therapeutic performance upon eliciting tumor cell apoptosis, which may cause chemoresistance of tumors. Design of novel drugs to eradicate apoptosis-resistant tumors via non-apoptotic cell death pathways is promising for improving the long-term chemotherapeutic efficacy. Herein, a Fe(III)-Shikonin metal-polyphenol-coordinated supramolecular nanomedicine for combined therapy of tumor via ferroptosis and necroptosis is designed. The construction of the nanomedicine based on the coordinated self-assembly between Fe3+ and Shikonin not only overcomes the shortcomings of Shikonin including its low bioavailability and high toxicity toward normal tissues, but also integrates the theranostics functions of Fe ions. Under the exposure of the high concentration of glutathione (GSH) in tumor cells, the as-prepared nanomedicine will disassemble into Fe2+ and Shikonin, followed by stimulating the tumor cell death through ferroptosis and necroptosis. In addition, benefiting from the stealth effect of polyethylene glycol (PEG) and the targeting ability of cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGD) to αv ß3 -integrin, NH2 -PEG-cRGD-modified nanomedicine exhibits a GSH-responsive therapy toward 4T1 tumor in vivo and self-enhanced longitudinal relaxation (T1 )-weighted imaging property. Since the self-assembly of natural Shikonin and human body-necessary Fe element is facile and feasible, the work may provide a promising supramolecular nanomedicine for next-generation chemotherapeutic applications.


Subject(s)
Ferroptosis , Cell Line, Tumor , Ferric Compounds , Humans , Nanomedicine , Naphthoquinones , Necroptosis
18.
Front Oncol ; 11: 778096, 2021.
Article in English | MEDLINE | ID: mdl-34900727

ABSTRACT

To investigate the effect of Lentinan (LNT) on lung adenocarcinoma (LUAD) cell stemness and its mechanism. In this study, we founded that LNT significantly reduce the cell proliferation, activity, migration, invasion, and stemness of LUAD cells, and promote their apoptosis compared with the control group in vitro. Moreover, LNT significantly inhibited the volume and weight of tumors of nude mice in vivo. At the same time, LNT can significantly up-regulate miR-216a-5p levels and reduce the protein expression of phospho-JAK2 (Y1007/1008) and phospho-STAT3 (Tyr705), thereby inhibiting the JAK2/STAT3 signaling pathway. Interfering with miR-216a-5p expression and activating the JAK2/STAT3 signaling pathway can significantly reverse LNT inhibitory effects on LUAD. Collectively, LNT can inhibit the JAK2/STAT3 signaling pathway by up-regulating miR-216a-5p, reducing stemness, and promoting LUAD cells apoptosis, then slow down LUAD occurrence and development, providing concepts and experimental foundation treating patients with LUAD.

19.
J Exp Clin Cancer Res ; 40(1): 278, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34470658

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM), a lethal brain tumor, remains the most daunting challenge in cancer therapy. Overexpression and constitutive activation of PDGFs and PDGFRα are observed in most GBM; however, available inhibitors targeting isolated signaling pathways are minimally effective. Therefore, better understanding of crucial mechanisms underlying GBM is needed for developing more effective targeted therapies. METHODS: Target genes controlled by HIF1α in GBM were identified by analysis of TCGA database and by RNA-sequencing of GBM cells with HIF1α knockout by sgRNA-Cas9 method. Functional roles of HIF1α, PDGFs and PDGFRs were elucidated by loss- or gain-of-function assays or chemical inhibitors, and compared in response to oxygen tension. Pharmacological efficacy and gene expression in mice with intracranial xenografts of primary GBM were analyzed by bioluminescence imaging and immunofluorescence. RESULTS: HIF1α binds the PDGFD proximal promoter and PDGFRA intron enhancers in GBM cells under normoxia or mild-hypoxia to induce their expression and maintain constitutive activation of AKT signaling, which in turn increases HIF1α protein level and activity. Paradoxically, severe hypoxia abrogates PDGFRα expression despite enhancing HIF1α accumulation and corresponding PDGF-D expression. Knockout of HIF1A, PDGFD or PDGFRA in U251 cells inhibits cell growth and invasion in vitro and eradicates tumor growth in vivo. HIF1A knockdown in primary GBM extends survival of xenograft mice, whereas PDGFD overexpression in GL261 shortens survival. HIF1α inhibitor Echinomycin induces GBM cell apoptosis and effectively inhibits growth of GBM in vivo by simultaneously targeting HIF1α-PDGFD/PDGFRα-AKT feedforward pathway. CONCLUSIONS: HIF1α orchestrates expression of PDGF-D and PDGFRα for constitutive activation of AKT pathway and is crucial for GBM malignancy. Therefore, therapies targeting HIF1α should provide an effective treatment for GBM.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Brain Neoplasms/pathology , Echinomycin/therapeutic use , Glioblastoma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Lymphokines/metabolism , Oxygen/metabolism , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Lymphokines/genetics , Mice , Mice, Inbred NOD , Neoplasm Invasiveness , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics
20.
Acta Pharmacol Sin ; 42(10): 1690-1702, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34112960

ABSTRACT

Ferroptotic cell death is characterized by iron-dependent lipid peroxidation that is initiated by ferrous iron and H2O2 via Fenton reaction, in which the role of activating transcription factor 3 (ATF3) remains elusive. Brucine is a weak alkaline indole alkaloid extracted from the seeds of Strychnos nux-vomica, which has shown potent antitumor activity against various tumors, including glioma. In this study, we showed that brucine inhibited glioma cell growth in vitro and in vivo, which was paralleled by nuclear translocation of ATF3, lipid peroxidation, and increases of iron and H2O2. Furthermore, brucine-induced lipid peroxidation was inhibited or exacerbated when intracellular iron was chelated by deferoxamine (500 µM) or improved by ferric ammonium citrate (500 µM). Suppression of lipid peroxidation with lipophilic antioxidants ferrostatin-1 (50 µM) or liproxstatin-1 (30 µM) rescued brucine-induced glioma cell death. Moreover, knockdown of ATF3 prevented brucine-induced accumulation of iron and H2O2 and glioma cell death. We revealed that brucine induced ATF3 upregulation and translocation into nuclei via activation of ER stress. ATF3 promoted brucine-induced H2O2 accumulation via upregulating NOX4 and SOD1 to generate H2O2 on one hand, and downregulating catalase and xCT to prevent H2O2 degradation on the other hand. H2O2 then contributed to brucine-triggered iron increase and transferrin receptor upregulation, as well as lipid peroxidation. This was further verified by treating glioma cells with exogenous H2O2 alone. Moreover, H2O2 reversely exacerbated brucine-induced ER stress. Taken together, ATF3 contributes to brucine-induced glioma cell ferroptosis via increasing H2O2 and iron.


Subject(s)
Activating Transcription Factor 3/metabolism , Antineoplastic Agents/therapeutic use , Ferroptosis/drug effects , Hydrogen Peroxide/metabolism , Iron/metabolism , Strychnine/analogs & derivatives , Amino Acid Transport System y+/metabolism , Animals , Antineoplastic Agents/pharmacology , Catalase/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , NADPH Oxidase 4/metabolism , Neoplasms/drug therapy , Strychnine/pharmacology , Strychnine/therapeutic use , Superoxide Dismutase-1/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...