Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Theranostics ; 12(13): 6038-6056, 2022.
Article in English | MEDLINE | ID: mdl-35966597

ABSTRACT

Rationale: Immunosuppression in the tumor microenvironment (TME) is key to the pathogenesis of solid tumors. Tumor cell-intrinsic autophagy is critical for sustaining both tumor cell metabolism and survival. However, the role of autophagy in the host immune system that allows cancer cells to escape immune destruction remains poorly understood. Here, we determined if attenuated host autophagy is sufficient to induce tumor rejection through reinforced adaptive immunity. Furthermore, we determined whether dietary glutamine supplementation, mimicking attenuated host autophagy, is capable of promoting antitumor immunity. Methods: A syngeneic orthotopic tumor model in Atg5+/+ and Atg5flox/flox mice was established to determine the impact of host autophagy on the antitumor effects against mouse malignant salivary gland tumors (MSTs). Multiple cohorts of immunocompetent mice were used for oncoimmunology studies, including inflammatory cytokine levels, macrophage, CD4+, and CD8+ cells tumor infiltration at 14 days and 28 days after MST inoculation. In vitro differentiation and in vivo dietary glutamine supplementation were used to assess the effects of glutamine on Treg differentiation and tumor expansion. Results: We showed that mice deficient in the essential autophagy gene, Atg5, rejected orthotopic allografts of isogenic MST cells. An enhanced antitumor immune response evidenced by reduction of both M1 and M2 macrophages, increased infiltration of CD8+ T cells, elevated IFN-γ production, as well as decreased inhibitory Tregs within TME and spleens of tumor-bearing Atg5flox/flox mice. Mechanistically, ATG5 deficiency increased glutamine level in tumors. We further demonstrated that dietary glutamine supplementation partially increased glutamine levels and restored potent antitumor responses in Atg5+/+ mice. Conclusions: Dietary glutamine supplementation exposes a previously undefined difference in plasticity between cancer cells, cytotoxic CD8+ T cells and Tregs.


Subject(s)
Glutamine , Salivary Gland Neoplasms , Animals , Autophagy , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , CD8-Positive T-Lymphocytes , Mice , Salivary Gland Neoplasms/drug therapy , Tumor Microenvironment
2.
Toxicol Appl Pharmacol ; 435: 115833, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34933056

ABSTRACT

Clinical utilization of doxorubicin (DOX), which is a commonly used chemotherapeutic, is restricted due to toxic effects on various tissues. Using hesperetin (HST), an antioxidant used in Chinese traditional medicine protects testis against DOX-induced toxicity although the molecular mechanisms are not well-known. The study was aimed to examine the possible role of the mechanistic target of rapamycin kinase (mTOR) and dynamin 1-like dynamin-related protein 1 (DRP1) in the therapeutic effects of HST on the DOX-induced testicular toxicity. Rats were divided into Control, DOX, DOX + HST, and HST groups (n = 7). Single-dose DOX (15 mg/kg) was administered intraperitoneally and HST (50 mg/kg) was administered by oral gavage every other day for 28 days. Total antioxidant status (TAS), histopathological evaluations, immunohistochemistry, and gene expression level detection analyses were performed. Histopathologically, DOX-induced testicular damage was ameliorated by HST treatment. DOX reduced testicular TAS levels and increased oxidative stress markers, 8-Hydroxy-deoxyguanosine (8-OHdG), and 4-Hydroxynonenal (4-HNE). Also, upregulated mTOR and DRP1 expressions with DOX exposure were decreased after HST treatment in the testis (p < 0.05). On the other hand, DOX-administration downregulated miR-150-5p and miR-181b-2-3p miRNAs, targeting mTOR and mRNA levels of beclin 1 (BECN1) and autophagy-related 5 (ATG5), autophagic markers. Furthermore, these levels were nearly similar to control testis samples in the DOX + HST group (p < 0.05). The study demonstrated that HST may have a therapeutic effect on DOX-induced testicular toxicity by removing reactive oxygen species (ROS) and by modulating the mTOR and DRP1 expressions, which have a critical role in regulating the balance of generation/elimination of ROS.


Subject(s)
Antibiotics, Antineoplastic , Doxorubicin , Dynamins/biosynthesis , Hesperidin/therapeutic use , TOR Serine-Threonine Kinases/biosynthesis , Testicular Diseases/chemically induced , Testicular Diseases/drug therapy , Animals , Antioxidants/metabolism , Autophagy-Related Protein 5/biosynthesis , Autophagy-Related Protein 5/genetics , Beclin-1/biosynthesis , Beclin-1/genetics , Dynamins/genetics , Gene Expression/drug effects , Male , MicroRNAs/biosynthesis , Oxidative Stress , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/genetics , Testicular Diseases/pathology , Testis/drug effects , Testis/metabolism , Testis/pathology
3.
Biomed Pharmacother ; 142: 112045, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34426257

ABSTRACT

OBJECTIVE: Asthma is characterized by airway hyperresponsiveness(AHR), inflammation and remodeling. Autophagy and endoplasmic reticulum stress(ERS) are dysregulated in asthma, and ATG5 has attracted wide attentions a representative gene of autophagy. Previous evidence shows that acupuncture may treat asthma by regulating the immune environment.However,the precise mechanism involved in acupuncture's effects on asthma is unclear. Thus, we investigated the inner-relationships of acupuncture and ATG5-mediated autophagy, ERS and CD4+ T lymphocyte differentiation in asthma. METHODS: Ovalbumin (OVA)-sensitized and challenged ATG5+/- and ATG5-/-mice with asthma were treated by acupuncture at Dazhui(GV14),Feishu(BL13) and Zusanli(ST36),and sacrificed the next day.Then blood and bronchoalveolar lavage fluid (BALF)samples were collected to determine inflammatory cell counts and cytokine levels. Lung tissue samples were obtained for histological examination, and the spleen was harvested for flow cytometry. RESULTS: Compared with the untreated group, acupuncture decreased BALF inflammatory cell counts and AHR in OVA-induced mice.Acupuncture decreased autophagy-related protein and mRNA (ATG5,Beclin-1,p62 and LC3B)amounts and ERS-related protein (p-PERK, p-IRE-1,Grp78, and ATF6)levels as well as autophagosome formation in lung tissue, concomitant with increased IFN-γ and decreased IL-4, IL-17 and TGF-ß amounts in BALF.Consistently, the imbalance of CD4+ T lymphocyte subsets(Th1/Th2 and Treg/Th17) was also corrected by acupuncture.Meanwhile, AHR and inflammation were decreased in ATG5-/- mice compared with ATG+/-animals,without affecting the therapeutic effect of acupuncture. CONCLUSION: Acupuncture reduces airway inflammation and AHR in asthma by inhibiting ATG5-mediated autophagy to regulate endoplasmic reticulum stress and CD4+T lymphocyte differentiation.


Subject(s)
Acupuncture Therapy , Asthma/therapy , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy/genetics , CD4-Positive T-Lymphocytes/immunology , Endoplasmic Reticulum Stress/genetics , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/pathology , Autophagosomes/ultrastructure , Autophagy/immunology , Autophagy-Related Protein 5/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cytokines/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress/immunology , Female , Inflammation/genetics , Inflammation/immunology , Mice, Inbred C57BL , Ovalbumin/toxicity , Respiratory Hypersensitivity
4.
J Tradit Chin Med ; 41(2): 236-245, 2021 04.
Article in English | MEDLINE | ID: mdl-33825403

ABSTRACT

OBJECTIVE: To explore the effects of Qingguang'an () containing serum on the expression levels of autophagy related genes in the transforming growth factor beta 1 (TGF-ß1)-activated human Tenon's fibroblasts (HTFs). METHODS: (a) Primary HTFs were stimulated by TGF-ß1 and underwent immunohistochemistry, which established a cell model after Glaucoma filtration surgery (GFS). (b) The cell models were divided into 4 group: normal group (normal cells), model group (+TGF-ß1),treatment group (+TGF-ß1+ medicated serum), and positive control group (TGF-ß1+ rapamycin). Then, Qingguang'an medicated serum with optimum concentration was added to the corresponding group. The autophagy positive cells were identified by the Cyto-ID autophagy detection kits under fluorescent microscope and Cytation 5 multifunctional instrument for cell imaging. And the mean fluorescence intensity of autophagy positive cells was determined by flow cytometry. The expression levels of autophagy related genes - Beclin-1, autophagy related gene 5 (ATG-5), and microtubule-associated protein 1 light chain 3 (LC-3Ⅱ were detected by quantitative reverse transcription-polymerase chain reaction and Western blot analysis. RESULTS: Compared with the normal group and the model group, the relative mRNA expression levels of autophagy-related genes (Beclin-1, ATG-5 and LC-3Ⅱ in the experimental group were notably increased (P < 0.05, P < 0.01), and with the extension of treatment time, it had an increasing trend (48 h was more obvious), which showed a certain time dependency; the protein expression levels of autophagy-related genes (Beclin-1, ATG-5, and LC-3Ⅱ were significantly increased in the experimental group (P < 0.05, P < 0.01). With the prolongation of treatment time, there was an increasing trend (48 h was relatively obvious), and it revealed a certain time dependency. CONCLUSION: The Qingguang'an medicated serum could up-regulate autophagy related genes (Beclin1, ATG5, and LC3Ⅱ in the TGF-ß1-activated HTFs.


Subject(s)
Autophagy-Related Protein 5/metabolism , Autophagy/drug effects , Beclin-1/metabolism , Fibroblasts/drug effects , Glaucoma/drug therapy , Tenon Capsule/drug effects , Transforming Growth Factor beta1/metabolism , Animals , Autophagy-Related Protein 5/genetics , Beclin-1/genetics , Cells, Cultured , Fibroblasts/metabolism , Glaucoma/genetics , Glaucoma/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley , Serum/chemistry , Tenon Capsule/cytology , Tenon Capsule/metabolism , Transforming Growth Factor beta1/genetics
5.
J Ethnopharmacol ; 270: 113821, 2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33460753

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Guizhi Fuling Wan (GFW) is a traditional Chinese medicine used to remove blood stasis and dissipate phlegm for treating gynecological diseases that was invented by Zhang Zhongjing in the Eastern Han dynasty. In recent years, GFW has been widely used to treat patients with polycystic ovary syndrome (PCOS). Clinical and animal studies have shown that it is effective in the treatment of PCOS, but its mechanism is unknown. Generally, it works by regulating autophagy via the PI3K/AKT/mTOR signaling pathway. AIM OF THE STUDY: This study investigated the effects and mechanism of GFW in PCOS rats with insulin resistance (IR) in order to provide better understanding of its observed clinical effects and a theoretical basis for the study of traditional Chinese medicine. MATERIALS AND METHODS: Eighty-four female Sprague-Dawley rats were randomly divided into seven groups (n = 12 per group): 1) control, 2) PCOS model, 3) low-dose GFW, 4) medium-dose GFW, 5) high-dose GFW, 6) metformin, and 7) medium-dose GFW plus LY294002. In all non-control groups, we induced PCOS through daily letrozole combined with intragastric high-fat emulsion for 21 days. After treatment, rats were sacrificed and serum follicle-stimulating hormone (FSH), testosterone (T), progesterone, luteinizing hormone (LH), 17ß-estradiol, fasting insulin (FINS), and fasting plasma glucose levels were measured by enzyme-linked immunosorbent assay (ELISA). The LH/FSH ratios and HOMA-IR values were calculated. Ovarian morphology was observed by hematoxylin and eosin staining, and all follicles were counted under a microscope. MDC-positive vesicles were used as markers to detect autophagy, and the expression levels of p62, Beclin1, and LC3-II were examined by immunostaining. Western blotting was used to measure PI3K/AKT/mTOR pathway activation, granulosa cell apoptosis, and autophagy. RESULTS: Compared with the PCOS model group, GFW-treated rats had less atretic and cystic follicles, and more mature follicles and corpus lutea. The GFW-treated rats had lower serum T, LH, and FINS levels than the PCOS model group, as well as lower LH/FSH ratios and HOMA-IR values. GFW treatment resulted in significantly reduced levels of cleaved-Caspase-3, cleaved-Caspase-9, BAX, Beclin1, Atg5, and LC3-II. Phosphorylation of PI3K, AKT, and mTOR was significantly higher in GFW-treated rats compared with the PCOS model group. The phosphorylation of PI3K, AKT, and mTOR was decreased with the use of a PI3K antagonist. CONCLUSIONS: Our results indicate that GFW inhibited granulosa cell autophagy and promoted follicular development to attenuate ovulation disorder in PCOS-IR rats. This was associated with activation of the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Autophagy/drug effects , Drugs, Chinese Herbal/pharmacology , Granulosa Cells/drug effects , Polycystic Ovary Syndrome/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Disease Models, Animal , Down-Regulation , Drugs, Chinese Herbal/therapeutic use , Female , Hormones/blood , Insulin Resistance , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Ovarian Follicle/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Polycystic Ovary Syndrome/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
6.
J Cell Physiol ; 236(2): 1148-1157, 2021 02.
Article in English | MEDLINE | ID: mdl-32686156

ABSTRACT

Saracatinib is an oral Src-kinase inhibitor and has been studied in preclinical models and clinical trials of cancer therapy. GMI, a fungal immunomodulatory protein from Ganoderma microsporum, possesses antitumor capacity. The aim of this study is to evaluate the cytotoxic effect of combination treatment with saracatinib and GMI on parental and pemetrexed-resistant lung cancer cells. Cotreatment with saracatinib and GMI induced synergistic and additive cytotoxic effect in A549 and A400 cells by annexin V/propidium iodide assay and combination index. Using western blot assay, saracatinib, and GMI combined treatment synergistically induced caspase-7 activation in A549 cells. Different from A549 cells, saracatinib and GMI cotreatment markedly increased LC3B-II in A400 cells. ATG5 silencing abolished the caspase-7 activation and reduced cell death in A549 cells after cotreatment. This is the first study to provide a novel strategy of treating lung cancer with or without drug resistance via combination treatment with GMI and saracatinib.


Subject(s)
Autophagy-Related Protein 5/genetics , Benzodioxoles/pharmacology , Caspase 7/genetics , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Quinazolines/pharmacology , src-Family Kinases/genetics , A549 Cells , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Cell Proliferation/drug effects , Fungal Proteins/chemistry , Fungal Proteins/pharmacology , Ganoderma/chemistry , Humans , Immunologic Factors/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Synthetic Lethal Mutations/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors
7.
Am J Chin Med ; 48(8): 1915-1940, 2020.
Article in English | MEDLINE | ID: mdl-33308096

ABSTRACT

Autophagy is an important tightly controlled cellular process that regulates cellular homeostasis and is involved in deciding cell fate such as cell survival and death. The role of autophagy in many intracellular signaling pathways explains its interaction with other different types of cell death, including apoptosis and immunogenic cell death (ICD). The reports showed the complex and intriguing relationship existing between autophagy and immune system signaling pathways. However, the role of autophagy in ICD remains to be clearly elucidated. In this study, we demonstrated that Brucine, a clinically-used small molecule in traditional Chinese medicine, elicited autophagy inhibition. Brucine also triggered cell stress and induced features of ICD, including calreticulin (CRT) exposure and high-mobility group box 1 (HMGB1) release in MDA-MB-231 and CT26 cancer cells. Brucine impaired autolysosomal degradation and exerted a feedback regulation of ERK1/2-mTOR-p70S6K signaling cascade. Brucine-elicited ICD was confirmed by the rejection of CT26 tumor cells, implanted in the mice after vaccination with Brucine-treated CT26 cells. The impaired autophagy contributed to Brucine-induced ICD, as knock-down of Atg5 significantly reduced Brucine-elicited CRT exposure and HMGB1 release. Our results revealed Brucine as a novel autophagy regulator, ICD inducer and hitherto undocumented role of autophagy in ICD. Thus, these results imply the importance of Brucine in cancer immunotherapy. Therefore, Brucine may be used as an ICD inducer and improve its application in cancer treatment with minimized toxicity.


Subject(s)
Autophagy/drug effects , Cell Death/genetics , Cell Death/immunology , Drugs, Chinese Herbal , Lysosomes/drug effects , Strychnine/analogs & derivatives , Animals , Autophagy/physiology , Autophagy-Related Protein 5/genetics , Calreticulin , Cell Line, Tumor , Gene Knockdown Techniques , HMGB1 Protein/metabolism , Humans , Immunotherapy , Lysosomes/physiology , MAP Kinase Signaling System/drug effects , Mice , Neoplasms/drug therapy , Phytotherapy , Strychnine/pharmacology , Strychnine/therapeutic use
8.
Mol Biol Rep ; 47(11): 9021-9032, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33034883

ABSTRACT

Despite the depth of knowledge concerning the pathogenesis of melanoma, the most aggressive type of skin cancer, the prognosis and survival of patients still remain a challenge. In addition, responses to chemotherapy and immunotherapy are still poor, which underscore an urgent need in the development of new therapeutic strategies for the treatment of melanoma. Recently, the dynamic role of autophagy has gained considerable interest in the pathogenesis and treatment of melanoma. Whereas a decrease in autophagy activity promotes melanoma formation by increasing oncogene-induced tumorigenesis and DNA damage accumulation, an enhanced level of autophagy sustains melanoma cell viability and contributes to drug resistance. Obviously, the understanding of autophagy regulation may lead to a better defining melanoma pathogenesis and the progression toward new treatment options. In this review, we present new insights into a dual role of autophagy during melanoma tumorigenesis. In addition to summarizing current therapeutics for treating melanoma, we discuss how autophagy manipulation may improve the patients' outcome. Finally, autophagy-modulating drugs and nanoparticles, alone or in combination with current therapeutics, are proposed for possible clinical use in melanoma management.


Subject(s)
Autophagy/physiology , Carcinogenesis/drug effects , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Sorafenib/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Carcinogenesis/genetics , Gene Expression/drug effects , Humans , Melanoma/genetics , Melanoma/physiopathology , Skin Neoplasms/genetics , Skin Neoplasms/physiopathology
9.
Am J Chin Med ; 48(4): 945-966, 2020.
Article in English | MEDLINE | ID: mdl-32476431

ABSTRACT

Tetramethylpyrazine has shown neuroprotective and axonal outgrowth-promoting effects and can improve cognitive deficit in a rat model of chronic hypoperfusion. However, the role of tetramethylpyrazine in sevoflurane-induced neurotoxicity is still vague. Therefore, this study was designed to investigate the effects and mechanisms of tetramethylpyrazine on sevoflurane-induced autophagy, apoptosis, and the expression of BACE1 and A[Formula: see text] in SH-SY5Y cells. We measured the expression levels of the apoptosis protein markers Bax and Bcl-2, autophagy protein markers Atg5 and LC3-II, BACE1, and A[Formula: see text] in SH-SY5Y cells after sevoflurane treatment and determined the effects of tetramethylpyrazine on sevoflurane-induced expression of these proteins after silencing GPR50 or Atg5 with siRNA in vitro. We found that exposure to 3.4% sevoflurane for 6 h decreased the expression of autophagy protein markers and increased the expression of the apoptosis protein markers, BACE1, and A[Formula: see text] in SH-SY5Y cells. The number of red puncta (autolysosomes) and yellow puncta (autophagosomes) in each SH-SY5Y cell decreased after transient transfection with the mRFP-GFP-LC3 expression plasmid. Silencing of GPR50 decreased the expression of pCREB, Atg5, and LC3-II, while silencing of Atg5 increased the expression of BACE1 and A[Formula: see text] in SH-SY5Y cells. Our results demonstrate that tetramethylpyrazine attenuated sevoflurane-induced neurotoxicity by enhancing autophagy through the GPR50/CREB pathway in SH-SY5Y cells.


Subject(s)
Autophagy/drug effects , Autophagy/genetics , CREB-Binding Protein/metabolism , Nerve Tissue Proteins/metabolism , Neuroprostanes , Pyrazines/pharmacology , Pyrazines/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Sevoflurane/toxicity , Signal Transduction/drug effects , Signal Transduction/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Gene Expression/drug effects , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Tumor Cells, Cultured
10.
Nat Cell Biol ; 22(4): 412-424, 2020 04.
Article in English | MEDLINE | ID: mdl-32203415

ABSTRACT

Although the transition metal copper (Cu) is an essential nutrient that is conventionally viewed as a static cofactor within enzyme active sites, a non-traditional role for Cu as a modulator of kinase signalling is emerging. Here, we found that Cu is required for the activity of the autophagic kinases ULK1 and ULK2 (ULK1/2) through a direct Cu-ULK1/2 interaction. Genetic loss of the Cu transporter Ctr1 or mutations in ULK1 that disrupt the binding of Cu reduced ULK1/2-dependent signalling and the formation of autophagosome complexes. Increased levels of intracellular Cu are associated with starvation-induced autophagy and are sufficient to enhance ULK1 kinase activity and, in turn, autophagic flux. The growth and survival of lung tumours driven by KRASG12D is diminished in the absence of Ctr1, is dependent on ULK1 Cu binding and is associated with reduced levels of autophagy and signalling. These findings suggest a molecular basis for exploiting Cu-chelation therapy to prevent autophagy signalling to limit proliferation and improve patient survival in cancer.


Subject(s)
Adenocarcinoma of Lung/genetics , Autophagy-Related Protein-1 Homolog/genetics , Autophagy/genetics , Copper/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/pathology , Amino Acid Sequence , Animals , Autophagosomes/enzymology , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Copper Transporter 1/deficiency , Copper Transporter 1/genetics , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/deficiency , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Xenograft Model Antitumor Assays
11.
Food Funct ; 10(12): 7634-7644, 2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31728459

ABSTRACT

Chemotherapy is currently used to treat colorectal cancer (CRC), the most common cancer worldwide. However, chemotherapeutic drugs are limited by severe side effects or drug resistance. In this study, bioactive compound(s), a mixture of palmitic acid, stearic acid, and glyceryl 1,3-dipalmitate (PSG), in Lactobacillus paracasei subsp. paracasei NTU 101-fermented reconstituted skimmed milk ethanol extract (NTU 101-FMEE) were isolated and identified. PSG (1 : 1.5 : 6.3) at 125 µg mL-1 could significantly decrease CRC cell viability at dosages that were not cytotoxic to healthy colon epithelial cells or macrophages. Moreover, the inhibitory effect of the combination of 62.5 µg mL-1 PSG (1 : 1.5 : 6.3) and 5-fluorouracil (5-FU) was significantly higher than that of 5-FU alone (p < 0.05). PSG up-regulated the activities of apoptosis-related proteins and down-regulated the nuclear factor-κB signaling pathway compared to the levels in the control group. Overall, PSG purified from NTU 101-FMEE possesses the potential to ameliorate CRC by improving the effects of adjuvant chemotherapy drugs and reducing side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Cultured Milk Products/analysis , Fluorouracil/pharmacology , Lacticaseibacillus paracasei/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Cultured Milk Products/microbiology , Fermentation , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Stearic Acids/metabolism , Stearic Acids/pharmacology
12.
Cell Physiol Biochem ; 53(5): 747-759, 2019.
Article in English | MEDLINE | ID: mdl-31622062

ABSTRACT

BACKGROUND/AIMS: Angiotensin II (Ang II) induces podocyte injury resulting in apoptosis in vitro and in vivo. However, the relationship between autophagy and apoptosis in Ang II-induced podocyte injury is unknown and the role of Ang II-induced autophagy in podocyte survival or death remains unclear. We investigated the sequential relationship between autophagy and apoptosis in Ang II-induced podocytes as well as the role of phosphatidylinositide 3-kinase (PI3-kinase). METHODS: Mouse podocytes were incubated in media containing various concentrations of Ang II and at different incubation times. The changes of podocyte autophagy and apoptosis were observed by electron microscopy, confocal imaging, western blotting, and FACS assay according to the presence of Ang II. RESULTS: Ang II enhanced the podocyte expression of the autophagic proteins, LC3A/B-II and beclin-1, and also increased the number of autophagosomes compared with control cells at early phase of 12 hours in a dose-dependent manner. This effect was inhibited by pretreatment with 3-methyladenine (3-MA), a PI3-kinase class III inhibitor. Thereafter, the Ang II-induced enhancement in autophagy decreased, whereas, podocyte apoptosis appeared later at 24 hours in concentration- and time-dependent manners in FACS and TUNEL assays. 3-MA and LY294002, a pan PI3-kinase inhibitor, further increased Ang II-induced podocyte apoptosis. Suppression of autophagy by Atg5 siRNA could induce podocyte apoptosis and further augment high-dose Ang II-induced podocyte apoptosis. CONCLUSION: These findings suggest that Ang II promotes autophagy in podocytes before apoptosis as an early adaptive cytoprotective mechanism for podocyte survival after Ang II treatment, and the transitional imbalance between autophagy and apoptosis causes podocyte injury.


Subject(s)
Angiotensin II/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagosomes/metabolism , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Beclin-1/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Podocytes/cytology , Podocytes/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation/drug effects
13.
BMC Complement Altern Med ; 19(1): 269, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31615565

ABSTRACT

BACKGROUND: Puerarin exerts therapeutic effect on osteoporosis due to its inhibitory effect on the formation of osteoclasts. Puerarin is also widely established as an autophagy inhibitor. The study aimed to investigate the significance of autophagy in Puerarin-treated osteoclast formation. METHODS: Osteoclast precursors (OCPs) derived from bone marrow-derived macrophages (BMMs) were treated with Puerarin along with RANKL or without RANKL, and then the autophagic parameters of OCPs (including autophagic proteins, LC3 transformation, autophagosome or LC3-puncta) were observed through Western Blotting, Transmission Electron Microscopy and Immunofluorescence assays. Next, after using overexpression vectors of autophagic genes (Atg7, Atg5 and BECN1) to alter autophagy activity, OCP proliferation was measured by Ethynyl deoxyuridine (EdU) assays and Cell Counting Kit-8 (CCK-8) kit, and osteoclast differentiation was assessed by Tartrate-resistant acid phosphatase (TRAP) staining. RESULTS: The results showed that Puerarin could directly inhibit the autophagy and proliferation of OCPs. Importantly, overexpression of autophagic genes Atg5, Atg7 and BECN1 reversed Puerarin-inhibited OCP autophagy and proliferation. What's more, RANKL could promote the autography of OCPs, which was recovered by Puerarin treatment. Interestingly, different from single-Puerarin treatment, we found that in the presence of RANKL, only BECN1 overexpression significantly reversed Puerarin-inhibited osteoclast differentiation and OCP autophagy. CONCLUSION: In conclusion, Puerarin could inhibit the OCP autophagy in the presence or absence of RANKL, which blocked the OCP proliferation and osteoclast differentiation respectively. Moreover, BECN1 plays an essential role in Puerarin-inhibited osteoclastogenesis. Our study provides potential clue to further complete the intrinsic mechanism of Puerarin in treating osteoporosis.


Subject(s)
Autophagy/drug effects , Drugs, Chinese Herbal/pharmacology , Isoflavones/pharmacology , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/metabolism , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Cells, Cultured , Female , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Osteoclasts/metabolism , Pueraria/chemistry , Signal Transduction/drug effects
14.
Food Funct ; 10(8): 5102-5114, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31363726

ABSTRACT

The present study was designed to investigate the protective effects of Cordyceps militaris polysaccharides (CMP) on STZ-treated DN mice. CMP were identified by FT-IR and HPLC. Diabetic nephropathy (DN) was induced in male C57BL/6 mice by the injection of streptozotocin (STZ, 50 mg kg-1) in citrate buffer on 5 consecutive days. Administration of CMP at 200 and 400 mg kg-1 or irbesartan at 60 mg kg-1 in the STZ-treated mice could prevent the damage caused by STZ. CMP significantly reduced the STZ-induced higher expression of the kidney index, TC, TG, MDA, urinary protein, Scr, and BUN, while it markedly increased the STZ-induced decrease in GSH levels compared with the DN group. Histopathology analysis of the kidney by PAS, Masson, and HE staining confirmed the renal injury induced by STZ and the protective effects of CMP. Transmission electron microscopy (TEM) results confirmed the severe foot process effacement induced by STZ, but CMP treatment inhibited the podocytes' structure defects and ameliorated the function of podocytes. Desmin was measured by immunofluorescence and was related to podocyte injury. The results showed that CMP lessened the expression of desmin induced by STZ. CD68 expression was measured by immunohistochemistry analysis, and the expressions of IL-1ß, IL-6, and MCP-1 mRNA were measured by qRT-PCR. The results showed that CMP suppressed the expressions of CD68, IL-1ß, IL-6, and MCP-1 mRNA induced by STZ. The role of autophagy in the treatment of DN mice with CMP was detected by TEM and western blotting. The results showed that the administration of CMP was able to overcome the STZ-treated autophagy deficiency, significantly increase the rate of autophagy in the kidney, promote the expression of Atg5, beclin1 and LC3 protein, and reduce the expression of p62 protein. In conclusion, the present study demonstrates that CMP exert a protective effect on DN in STZ-treated mice possibly via activation of autophagy.


Subject(s)
Cordyceps/chemistry , Diabetic Nephropathies/drug therapy , Plant Extracts/administration & dosage , Polysaccharides/administration & dosage , Animals , Autophagy/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL
15.
Breast Cancer ; 26(6): 766-775, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31172425

ABSTRACT

BACKGROUND: Icariin is a major component isolated from Epimedium brevicornum Maxim and has been reported to exhibit anti-tumor activity. However, whether icariin could reverse the acquired drug resistance in breast cancer remains largely unclear. Therefore, this study was designed to explore the antitumor effects of icariin and its underlying mechanisms in a tamoxifen-resistant breast cancer cell line MCF-7/TAM. METHODS: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and Lactate dehydrogenase (LDH) assay were performed to determine the effects of icariin on cell viability and cell death. Cell cycle progression and apoptosis were detected by flow cytometry analysis. Transmission electron microscopy (TEM) assay was utilized to observe cell autophagy. The downstream protein levels were measured using western blotting. RESULTS: Here, we observed that icariin treatment not only inhibited the growth of MCF-7 but also has a potential function to overcome tamoxifen resistance in MCF-7/TAM. Moreover, icariin significantly induced cell cycle G0/G1 phase arrest and apoptosis, as well as suppressed autophagy. At molecular levels, icariin treatment remarkably down-regulated the expression levels of CDK2, CDK4, Cyclin D1, Bcl-2, LC3-1, LC3-II, AGT5, Beclin-1, but upregulated the expression levels of caspase-3, PARP and p62. Most importantly, we found inhibition of autophagy via 3-MA treatment could significantly enhance the effects of icariin on cell viability and apoptosis. Enhanced autophagy via autophagy related 5 (ATG5) overexpression could partially reverse the effects of icariin on cell viability and apoptosis. CONCLUSION: These results revealed that icariin might potentially be useful as an adjuvant agent in cancer chemotherapy to enhance the effect of tamoxifen through suppression of autophagy in vitro and provide insight into the therapeutic potential of icariin for the treatment of chemo-resistant breast cancer.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , Plant Extracts/pharmacology , Tamoxifen/adverse effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Breast Neoplasms/drug therapy , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Epimedium/chemistry , Female , Humans , MCF-7 Cells , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/therapeutic use , Signal Transduction/drug effects , Tamoxifen/therapeutic use , Transfection
16.
Nature ; 563(7732): 569-573, 2018 11.
Article in English | MEDLINE | ID: mdl-30429607

ABSTRACT

Autophagy captures intracellular components and delivers them to lysosomes, where they are degraded and recycled to sustain metabolism and to enable survival during starvation1-5. Acute, whole-body deletion of the essential autophagy gene Atg7 in adult mice causes a systemic metabolic defect that manifests as starvation intolerance and gradual loss of white adipose tissue, liver glycogen and muscle mass1. Cancer cells also benefit from autophagy. Deletion of essential autophagy genes impairs the metabolism, proliferation, survival and malignancy of spontaneous tumours in models of autochthonous cancer6,7. Acute, systemic deletion of Atg7 or acute, systemic expression of a dominant-negative ATG4b in mice induces greater regression of KRAS-driven cancers than does tumour-specific autophagy deletion, which suggests that host autophagy promotes tumour growth1,8. Here we show that host-specific deletion of Atg7 impairs the growth of multiple allografted tumours, although not all tumour lines were sensitive to host autophagy status. Loss of autophagy in the host was associated with a reduction in circulating arginine, and the sensitive tumour cell lines were arginine auxotrophs owing to the lack of expression of the enzyme argininosuccinate synthase 1. Serum proteomic analysis identified the arginine-degrading enzyme arginase I (ARG1) in the circulation of Atg7-deficient hosts, and in vivo arginine metabolic tracing demonstrated that serum arginine was degraded to ornithine. ARG1 is predominantly expressed in the liver and can be released from hepatocytes into the circulation. Liver-specific deletion of Atg7 produced circulating ARG1, and reduced both serum arginine and tumour growth. Deletion of Atg5 in the host similarly regulated [corrected] circulating arginine and suppressed tumorigenesis, which demonstrates that this phenotype is specific to autophagy function rather than to deletion of Atg7. Dietary supplementation of Atg7-deficient hosts with arginine partially restored levels of circulating arginine and tumour growth. Thus, defective autophagy in the host leads to the release of ARG1 from the liver and the degradation of circulating arginine, which is essential for tumour growth; this identifies a metabolic vulnerability of cancer.


Subject(s)
Arginine/blood , Autophagy , Neoplasms/blood , Neoplasms/pathology , Allografts , Animals , Arginase/blood , Arginase/metabolism , Arginine/administration & dosage , Arginine/pharmacology , Autophagy/genetics , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dietary Supplements , Hepatocytes/enzymology , Hepatocytes/metabolism , Liver/enzymology , Male , Mice , Neoplasm Transplantation , Neoplasms/genetics , Ornithine/metabolism
17.
Food Funct ; 9(11): 5682-5696, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30310905

ABSTRACT

Numerous studies propose that epigallocatechin-3-gallate (EGCG), an abundant polyphenol in green tea, has anti-cancer properties. However, its mechanism of action in breast cancer remains unclear. This study investigated the capacity of EGCG to suppress breast cancer cell growth in vitro and in vivo, characterizing the underlying mechanisms, focusing on the effect of EGCG on glucose metabolism. EGCG reduced breast cancer 4T1 cell growth in a concentration- (10-320 µM) and time- (12-48 h) dependent manner. EGCG induced breast cancer apoptotic cell death at 24 h, as evidenced by annexin V/PI, caspase 3, caspase 8 and caspase 9 activation. Furthermore, EGCG affected the expression of 16 apoptosis-related genes, and promoted mitochondrial depolarization. EGCG induced autophagy concentration-dependently in 4T1 cells by modulating the levels of the autophagy-related proteins Beclin1, ATG5 and LC3B. Moreover, EGCG affected glucose, lactate and ATP levels. Mechanistically, EGCG significantly inhibited the activities and mRNA levels of the glycolytic enzymes hexokinase (HK), phosphofructokinase (PFK), and lactic dehydrogenase (LDH), and to a lesser extent the activity of pyruvate kinase (PK). In addition, EGCG decreased the expression of hypoxia-inducible factor 1α (HIF1α) and glucose transporter 1 (GLUT1), critical players in regulating glycolysis. In vivo, EGCG reduced breast tumor weight in a dose-dependent manner, reduced glucose and lactic acid levels and reduced the expression of the vascular endothelial growth factor (VEGF). In conclusion, EGCG exerts an anti-tumor effect through the inhibition of key enzymes that participate in the glycolytic pathway and the suppression of glucose metabolism.


Subject(s)
Breast Neoplasms/drug therapy , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Glucose/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Catechin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Polyphenols/pharmacology , Tea/chemistry , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
18.
Sci Rep ; 8(1): 10348, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29985441

ABSTRACT

Glioblastoma Multiforme (GBM) is known to be one of the most malignant and aggressive forms of brain cancer due to its resistance to chemotherapy. Recently, GBM was found to not only utilise both oxidative phosphorylation (OXPHOS) and aerobic glycolysis, but also depend on the bulk protein degradation system known as macroautophagy to uphold proliferation. Although autophagy modulators hold great potential as adjuvants to chemotherapy, the degree of upregulation or inhibition necessary to achieve cell death sensitisation remains unknown. Therefore, this study aimed to determine the degree of autophagy modulation necessary to impair mitochondrial bioenergetics to the extent of promoting cell death onset. It was shown that coordinated upregulation of autophagy followed by its inhibition prior to chemotherapy decreased electron transfer system (ETS) and oxidative phosphorylation (OXPHOS) capacity, impaired mitochondrial fission and fusion dynamics and enhanced apoptotic cell death onset in terms of cleaved caspase 3 and cleaved PARP expression. Therefore, coordinated autophagy modulation may present a favourable avenue for improved chemotherapeutic intervention in the future.


Subject(s)
Autophagy , Drug Resistance, Neoplasm , Mitochondria/metabolism , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Electron Transport Complex I/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Lactic Acid/metabolism , Mitochondrial Dynamics/drug effects , Oxidative Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Temozolomide/pharmacology
19.
Front Immunol ; 9: 269, 2018.
Article in English | MEDLINE | ID: mdl-29515581

ABSTRACT

Interleukin-1ß (IL-1ß), a major pro-inflammatory cytokine, is a leaderless cytosolic protein whose secretion does not follow the classical endoplasmic reticulum-to-Golgi pathway, and for which a canonical mechanism of secretion remains to be established. Neutrophils are essential players against bacterial and fungi infections. These cells are rapidly and massively recruited from the circulation into infected tissues and, beyond of displaying an impressive arsenal of toxic weapons effective to kill pathogens, are also an important source of IL-1ß in infectious conditions. Here, we analyzed if an unconventional secretory autophagy mechanism is involved in the exportation of IL-1ß by these cells. Our findings indicated that inhibition of autophagy with 3-methyladenine and Wortmannin markedly reduced IL-1ß secretion induced by LPS + ATP, as did the disruption of the autophagic flux with Bafilomycin A1 and E64d. These compounds did not noticeable affect neutrophil viability ruling out that the effects on IL-1ß secretion were due to cell death. Furthermore, VPS34IN-1, a specific autophagy inhibitor, was still able to reduce IL-1ß secretion when added after it was synthesized. Moreover, siRNA-mediated knockdown of ATG5 markedly reduced IL-1ß secretion in neutrophil-differentiated PLB985 cells. Upon LPS + ATP stimulation, IL-1ß was incorporated to an autophagic compartment, as was revealed by its colocalization with LC3B by confocal microscopy. Overlapping of IL-1ß-LC3B in a vesicular compartment peaked before IL-1ß increased in culture supernatants. On the other hand, stimulation of autophagy by cell starvation augmented the colocalization of IL-1ß and LC3B and then promoted neutrophil IL-1ß secretion. In addition, specific ELISAs indicated that although both IL-1ß and pro-IL-1ß are released to culture supernatants upon neutrophil stimulation, autophagy only promotes IL-1ß secretion. Furthermore, the serine proteases inhibitor AEBSF reduced IL-1ß secretion. Moreover, IL-1ß could be also found colocalizing with elastase, suggesting both some vesicles containing IL-1ß intersect azurophil granules content and that serine proteases also regulate IL-1ß secretion. Altogether, our findings indicate that an unconventional autophagy-mediated secretory pathway mediates IL-1ß secretion in human neutrophils.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Neutrophils/immunology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine Triphosphate/immunology , Autophagy/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Cell Line , Humans , Lipopolysaccharides/immunology , Macrolides/pharmacology , Microtubule-Associated Proteins/metabolism , Protein Transport , RNA, Small Interfering/genetics , Secretory Pathway , Serine Proteases/metabolism , Wortmannin/pharmacology
20.
Phytomedicine ; 39: 160-167, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29433677

ABSTRACT

BACKGROUND: Lung adenocarcinoma is the most primary histologic subtype of non-small cell lung cancer (NSCLC). Compound 8b, a novel coumarin derivative with phenylsulfonylfuroxan group, shows significant antiproliferation activity against lung adenocarcinoma cell with low toxicity. PURPOSE: This study aims to uncover the potential of compound 8b in relation to apoptosis as well as autophagy induction in lung adenocarcinoma cells. STUDY DESIGN: The cytotoxicity and apoptosis of A549 and H1299 cells induced by compound 8b were detected by MTT, microscope and western blot analysis. Autophagy was determined by TEM, confocal microscopy and western blot analysis. Akt/mTOR and Erk signaling pathway were also examined by western blot analysis. RESULTS: First, significant growth inhibition and caspase-dependent apoptosis were observed in compound 8b-treated A549 and H1299 cells. Then, we confirmed compound 8b-induced autophagy by autophagosomes formation, upregulated expression of autophagy-related protein LC3-II and autophagic flux. Importantly, abolishing autophagy using inhibitors and ATG5 siRNA enhanced the cytotoxicity of compound 8b, indicating the cytoprotective role of autophagy in lung adenocarcinoma. Further mechanistic investigations suggested that Akt/mTOR and Erk signaling pathways contributed to autophagy induction by compound 8b. CONCLUSION: This results demonstrate that compound 8b induces caspase-dependent apoptosis as well as cytoprotective autophagy in lung adenocarcinoma cells, which may provide scientific evidence for developing this furoxan-based NO-releasing coumarin derivative as a potential anti-lung adenocarcinoma therapeutic agents.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Lung Neoplasms/drug therapy , Oxadiazoles/chemistry , A549 Cells , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Apoptosis/drug effects , Apoptosis/physiology , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Caspases/metabolism , Cell Line, Tumor , Coumarins/chemistry , Humans , Lung Neoplasms/pathology , Nitric Oxide/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL