1
|
Tedesco G, Santarosa M, Maestro R. Beyond self‑eating: Emerging autophagy‑independent functions for the autophagy molecules in cancer (Review). Int J Oncol 2024; 64:57. [PMID: 38606507 PMCID: PMC11087037 DOI: 10.3892/ijo.2024.5645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Autophagy is a conserved catabolic process that controls organelle quality, removes misfolded or abnormally aggregated proteins and is part of the defense mechanisms against intracellular pathogens. Autophagy contributes to the suppression of tumor initiation by promoting genome stability, cellular integrity, redox balance and proteostasis. On the other hand, once a tumor is established, autophagy can support cancer cell survival and promote epithelial‑to‑mesenchymal transition. A growing number of molecules involved in autophagy have been identified. In addition to their key canonical activity, several of these molecules, such as ATG5, ATG12 and Beclin‑1, also exert autophagy‑independent functions in a variety of biological processes. The present review aimed to summarize autophagy‑independent functions of molecules of the autophagy machinery and how the activity of these molecules can influence signaling pathways that are deregulated in cancer progression.
Collapse
Affiliation(s)
- Giulia Tedesco
- Unit of Oncogenetics and Functional Oncogenomics, CRO Aviano, National Cancer Institute, IRCCS, I-33081 Aviano, Italy
| | - Manuela Santarosa
- Unit of Oncogenetics and Functional Oncogenomics, CRO Aviano, National Cancer Institute, IRCCS, I-33081 Aviano, Italy
| | - Roberta Maestro
- Unit of Oncogenetics and Functional Oncogenomics, CRO Aviano, National Cancer Institute, IRCCS, I-33081 Aviano, Italy
| |
Collapse
|
2
|
Nokhostin F, Azadehrah M, Azadehrah M. The multifaced role and therapeutic regulation of autophagy in ovarian cancer. Clin Transl Oncol 2022; 25:1207-1217. [PMID: 36534371 DOI: 10.1007/s12094-022-03045-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Ovarian cancer (OC) is one of the tumors that occurs most frequently in women. Autophagy is involved in cell homeostasis, biomolecule recycling, and survival, making it a potential target for anti-tumor drugs. It is worth noting that growing evidence reveals a close link between autophagy and OC. In the context of OC, autophagy demonstrates activity as both a tumor suppressor and a tumor promoter, depending on the context. Autophagy's exact function in OC is greatly reliant on the tumor microenvironment (TME) and other conditions, such as hypoxia, nutritional deficiency, chemotherapy, and so on. However, what can be concluded from different studies is that autophagy-related signaling pathways, especially PI3K/AKT/mTOR axis, increase in advanced stages and malignant phenotype of the disease reduces autophagy and ultimately leads to tumor progression. This study sought to present a thorough understanding of the role of autophagy-related signaling pathways in OC and existing therapies targeting these signaling pathways.
Collapse
Affiliation(s)
- Fahimeh Nokhostin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Mahboobeh Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Malihe Azadehrah
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
3
|
Shi M, Maique J, Shepard S, Li P, Seli O, Moe OW, Chang Hu M. In vivo evidence for therapeutic applications of beclin 1 to promote recovery and inhibit fibrosis after acute kidney injury. Kidney Int 2022; 101:63-78. [PMID: 34736972 PMCID: PMC8741729 DOI: 10.1016/j.kint.2021.09.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 01/18/2023]
Abstract
Autophagy regulator beclin 1 activity determines the severity of kidney damage induced by ischemia reperfusion injury, but its role in kidney recovery and fibrosis are unknown and its therapeutic potentials have not been tested. Here, we explored beclin 1 effects on kidney fibrosis in three models of acute kidney injury (AKI)-ischemia reperfusion injury, cisplatin kidney toxicity, and unilateral ureteric obstruction in mouse strains with three levels of beclin 1 function: normal (wild type), low (heterozygous global deletion of beclin 1, Becn1+/-), and high beclin 1 activity (knockin gain-of-function mutant Becn1, Becn1FA). Fourteen days after AKI induction, heterozygous mice had more, but knockin mice had less kidney fibrosis than wild-type mice did. One day after ischemia reperfusion injury, heterozygous pan-kidney tubular Becn1 null mice had more severe kidney damage than homozygous distal tubular Becn1 null mice did, which was similar to the wild-type mice, implying that proximal tubular beclin 1 protects the kidney against ischemia reperfusion injury. By 14 days, both pan-kidney heterozygous Becn1 null and distal tubular homozygous Becn1 null mice had poorer kidney recovery than wild-type mice did. Injection of beclin 1 peptides increased cell proliferation in kidney tubules in normal mice. Beclin 1 peptides injection either before or after (2-5 days) ischemia reperfusion injury protected the kidney from injury and suppressed kidney fibrosis. Thus, both endogenous beclin 1 protein expression in kidney tubules and exogenous beclin 1 peptides are kidney protective via attenuation of acute kidney damage, promotion of cell proliferation, and inhibition of kidney fibrosis, consequently improving kidney recovery post-AKI. Hence, exogenous beclin 1 peptide may be a potential new therapy for AKI.
Collapse
Affiliation(s)
- Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Sierra Shepard
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Peng Li
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Olivia Seli
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Orson W. Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA,Address for reprint request and other correspondence: Ming Chang Hu, MD, PhD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA, or Orson W. Moe, MD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA,
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA,Address for reprint request and other correspondence: Ming Chang Hu, MD, PhD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA, or Orson W. Moe, MD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA,
| |
Collapse
|
4
|
MiR-200c sensitizes Olaparib-resistant ovarian cancer cells by targeting Neuropilin 1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:3. [PMID: 31898520 PMCID: PMC6939329 DOI: 10.1186/s13046-019-1490-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ovarian cancer (OC) is the most lethal gynecological malignancy and the second leading cause of cancer-related death in women. Treatment with PARP inhibitors (PARPi), such as Olaparib, has been recently introduced for OC patients, but resistance may occur and underlying mechanisms are still poorly understood. The aim of this study is to identify target genes within the tumor cells that might cause resistance to Olaparib. We focused on Neuropilin 1 (NRP1), a transmembrane receptor expressed in OC and correlated with poor survival, which has been also proposed as a key molecule in OC multidrug resistance. METHODS Using three OC cell lines (UWB, UWB-BRCA and SKOV3) as model systems, we evaluated the biological and molecular effects of Olaparib on OC cell growth, cell cycle, DNA damage and apoptosis/autophagy induction, through MTT and colony forming assays, flow cytometry, immunofluorescence and Western blot analyses. We evaluated NRP1 expression in OC specimens and cell lines by Western blot and qRT-PCR, and used RNA interference to selectively inhibit NRP1. To identify miR-200c as a regulator of NRP1, we used miRNA target prediction algorithms and Pearsons' correlation analysis in biopsies from OC patients. Then, we used a stable transfection approach to overexpress miR-200c in Olaparib-resistant cells. RESULTS We observed that NRP1 is expressed at high levels in resistant cells (SKOV3) and is upmodulated in partially sensitive cells (UWB-BRCA) upon prolonged Olaparib treatment, leading to poor drug response. Our results show that the selective inhibition of NRP1 is able to overcome Olaparib resistance in SKOV3 cells. Moreover, we demonstrated that miR-200c can target NRP1 in OC cells, causing its downmodulation, and that miR-200c overexpression is a valid approach to restore Olaparib sensitivity in OC resistant cells. CONCLUSIONS These data demonstrate that miR-200c significantly enhanced the anti-cancer efficacy of Olaparib in drug-resistant OC cells. Thus, the combination of Olaparib with miRNA-based therapy may represent a promising treatment for drug resistant OC, and our data may help in designing novel precision medicine trials for optimizing the clinical use of PARPi.
Collapse
|
5
|
Pan H, Wang Y, Na K, Wang Y, Wang L, Li Z, Guo C, Guo D, Wang X. Autophagic flux disruption contributes to Ganoderma lucidum polysaccharide-induced apoptosis in human colorectal cancer cells via MAPK/ERK activation. Cell Death Dis 2019; 10:456. [PMID: 31186406 PMCID: PMC6560101 DOI: 10.1038/s41419-019-1653-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 01/02/2023]
Abstract
Targeting autophagy may serve as a promising strategy for cancer therapy. Ganoderma lucidum polysaccharide (GLP) has been shown to exert promising anti-cancer effects. However, the underlying mechanisms remain elusive. Whether GLP regulates autophagy in cancer has never been reported. In this study, GLP induced the initiation of autophagy in colorectal cancer (CRC) HT-29 and HCT116 cells, as evidenced by enhanced level of LC3-II protein, GFP-LC3 puncta, and increased formation of double membrane vacuoles. However, GLP treatment caused marked increase of p62 expression. Addition of late stage autophagy inhibitor, chloroquine (CQ), further enhanced LC3-II and p62 level, as well as increased autophagosome accumulation, suggesting a blockage of autophagic flux by GLP in CRC cells. We then found GLP blocked autophagosome and lysosome fusion as determined by mRFP-GFP-LC3 colocalization analysis. Mechanistic study revealed that GLP-induced disruption of autophagosome-lysosome fusion is due to reduced lysosome acidification and lysosomal cathepsin activities. Cell viability and flow cytometry assays revealed that GLP-induced autophagosome accumulation is responsible for GLP-induced apoptosis in CRC cells. In line with this, inhibition of autophagy initiation by 3-methyladenine (3-MA), an early stage autophagy inhibitor, attenuated GLP-induced apoptosis. In contrast, suppression of autophagy at late stage by CQ enhanced the anti-cancer effect of GLP. Furthermore, we demonstrated that GLP-induced autophagosome accumulation and apoptosis is mediated via MAPK/ERK activation. Finally, GLP inhibited tumor growth and also inhibited autophagic flux in vivo. These results unveil new molecular mechanism underlying anti-cancer effects of GLP, suggesting that GLP is a potent autophagy inhibitor and might be useful in anticancer therapy.
Collapse
Affiliation(s)
- Haitao Pan
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Yujie Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Kun Na
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Ying Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Lu Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Zhenhao Li
- Zhejiang Shouxiangu Institute of Rare Medicine Plant, 12, Huanglong 3rd Road, 321200, Wuyi, Zhejiang, China
| | - Chengjie Guo
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Dandan Guo
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China
| | - Xingya Wang
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, 310053, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Zhu J, Cai Y, Xu K, Ren X, Sun J, Lu S, Chen J, Xu P. Beclin1 overexpression suppresses tumor cell proliferation and survival via an autophagy‑dependent pathway in human synovial sarcoma cells. Oncol Rep 2018; 40:1927-1936. [PMID: 30066884 PMCID: PMC6111547 DOI: 10.3892/or.2018.6599] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022] Open
Abstract
Beclin1 is an important autophagy‑related prot-ein, which is involved in both autophagy and apoptosis. In recent years, the antitumor effect of Beclin1 has received increased attention. In the present study, we established a stable Beclin1‑overexpressing cell line with SW982 human synovial sarcoma cells. We found that Beclin1 overexpression decreased the cell viability, inhibited proliferation and induced apoptosis in SW982 cells. The expression levels of Bcl‑2 and PCNA were decreased, while the levels of cleaved‑caspase‑3 and cleaved‑PARP were increased. Beclin1 is closely related with autophagy, thus the autophagy‑related markers LC3 and p62 were detected by western blot analysis, and transmission electron microscopy was used to observe autophagosomes. The results showed that the expression level of LC3II was increased and that of p62 was decreased. Moreover, many double membrane‑enclosed autophagosomes were found in cells with Beclin1 overexpression, which indicated that the autophagic activity was enhanced. To explore the effect of autophagy on the viability of SW982 cells, Atg5 was knocked down using siRNA to inhibit the autophagic activity. We found that autophagy contributed to the decrease in cell viability. Knockdown of Atg5 increased the viability and decreased the apoptotic rate of SW982 cells with Beclin1 overexpression. The expression level of Bcl‑2 was increased, while the expression levels of cleaved‑caspase‑3 and cleaved‑PARP were decreased. We also found that the Akt/Bcl‑2/caspase‑9 pathway was involved. The phosphorylation of AKT was positively correlated with cell viability. The cleavage of caspase‑9 was increased by Beclin1 overexpression and decreased by inhibition of autophagy. Altogether, our results suggested that both autophagy and apoptosis contributed to the antitumor effect of Beclin1 in SW982 cells.
Collapse
Affiliation(s)
- Jialin Zhu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Yongsong Cai
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Ke Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaoyu Ren
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Jian Sun
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Shemin Lu
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Jinghong Chen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Institute of Endemic Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
7
|
Zhan L, Zhang Y, Wang W, Song E, Fan Y, Li J, Wei B. Autophagy as an emerging therapy target for ovarian carcinoma. Oncotarget 2018; 7:83476-83487. [PMID: 27825125 PMCID: PMC5347782 DOI: 10.18632/oncotarget.13080] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/21/2016] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a conserved cellular self-digestion pathway for maintenance of homeostasis under basal and stressed conditions. Autophagy plays pivotal roles in the pathogenesis of many diseases, such as aging-related diseases, autoimmune diseases, cardiovascular diseases, and cancers. Of special note is that accumulating data suggest an intimate relationship between autophagy and ovarian carcinoma. Autophagy is well identified to act as either as a tumor-suppressor or as a tumor-promoter in ovarian carcinoma. The exact function of autophagy in ovarian carcinoma is highly dependent on the circumstances of cancer including hypoxic, nutrient-deficient, chemotherapy and so on. However, the mechanism underlying autophagy associated with ovarian carcinoma remains elusive, the precise role of autophagy in ovarian carcinoma also remains undetermined. In this review, we tried to sum up and discuss recent research achievements of autophagy in ovarian cancer. Moreover, waves of novel therapies ways for ovarian carcinoma based on the functions of autophagy were collected.
Collapse
Affiliation(s)
- Lei Zhan
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yu Zhang
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wenyan Wang
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Enxue Song
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yijun Fan
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China
| | - Bing Wei
- Department of gynecology and obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
8
|
Kalai Selvi S, Vinoth A, Varadharajan T, Weng CF, Vijaya Padma V. Neferine augments therapeutic efficacy of cisplatin through ROS- mediated non-canonical autophagy in human lung adenocarcinoma (A549 cells). Food Chem Toxicol 2017; 103:28-40. [DOI: 10.1016/j.fct.2017.02.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 01/25/2017] [Accepted: 02/14/2017] [Indexed: 12/31/2022]
|
9
|
Sok SPM, Arshad NM, Azmi MN, Awang K, Ozpolat B, Hasima Nagoor N. The apoptotic effect of 1'S-1'-Acetoxychavicol Acetate (ACA) enhanced by inhibition of non-canonical autophagy in human non-small cell lung cancer cells. PLoS One 2017; 12:e0171329. [PMID: 28158287 PMCID: PMC5291426 DOI: 10.1371/journal.pone.0171329] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
Abstract
Autophagy plays a role in deciding the fate of cells by inducing either survival or death. 1’S-1-acetoxychavicol acetate (ACA) is a phenylpropanoid isolated from rhizomes of Alpinia conchigera and has been reported previously on its apoptotic effects on various cancers. However, the effect of ACA on autophagy remains ambiguous. The aims of this study were to investigate the autophagy-inducing ability of ACA in human non-small cell lung cancer (NSCLC), and to determine its role as pro-survival or pro-death mechanism. Cell viability assay was conducted using MTT. The effect of autophagy was assessed by acridine orange staining, GFP-LC3 punctate formation assay, and protein level were analysed using western blot. Annexin V-FITC/PI staining was performed to detect percentage of cells undergoing apoptosis by using flow cytometry. ACA inhibits the cell viability and induced formation of cytoplasmic vacuoles in NSCLC cells. Acidic vesicular organelles and GFP-LC3 punctate formation were increased in response to ACA exposure in A549 and SK-LU-1 cell lines; implying occurrence of autophagy. In western blot, accumulation of LC3-II accompanied by degradation of p62 was observed, which further confirmed the full flux of autophagy induction by ACA. The reduction of Beclin-1 upon ACA treatment indicated the Beclin-1-independent autophagy pathway. An early autophagy inhibitor, 3-methyaldenine (3-MA), failed to suppress the autophagy triggered by ACA; validating the existence of Beclin-1-independent autophagy. Silencing of LC3-II using short interfering RNA (siRNA) abolished the autophagy effects, enhancing the cytotoxicity of ACA through apoptosis. This proposed ACA triggered a pro-survival autophagy in NSCLC cells. Consistently, co-treatment with lysosomal inhibitor, chloroquine (CQ), exerted a synergistic effect resulting in apoptosis. Our findings suggested ACA induced pro-survival autophagy through Beclin-1-independent pathway in NSCLC. Hence, targeting autophagy pathway using autophagy inhibitor such as CQ represented a novel promising approach to potentiate the cytotoxicity of ACA through apoptosis in NSCLC.
Collapse
Affiliation(s)
- Sophia P. M. Sok
- Institute of Biological Sciences (Genetics and Molecular Biology), Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Norhafiza M. Arshad
- Institute of Biological Sciences (Genetics and Molecular Biology), Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- Centre for Research in Biotechnology for Agriculture (CEBAR), University of Malaya, Kuala Lumpur, Malaysia
| | - Mohamad Nurul Azmi
- Centre of Natural Product Research and Drug Discovery (CENAR), Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Khalijah Awang
- Centre of Natural Product Research and Drug Discovery (CENAR), Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Noor Hasima Nagoor
- Institute of Biological Sciences (Genetics and Molecular Biology), Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- Centre for Research in Biotechnology for Agriculture (CEBAR), University of Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
10
|
Hu Z, Cai M, Deng L, Zhu L, Gao J, Tan M, Liu J, Lin B. The fucosylated CD147 enhances the autophagy in epithelial ovarian cancer cells. Oncotarget 2016; 7:82921-82932. [PMID: 27863372 PMCID: PMC5347741 DOI: 10.18632/oncotarget.13289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/17/2016] [Indexed: 11/29/2022] Open
Abstract
Autophagy is modulated by multiple factors including CD147, but little is know about the effects and mechanism by which the modification of CD147 by Lewis y antigen regulates autophagy of ovarian cancer cell. Here, we reported that Lewis y antigen can promote basic autophagy activity and restrain autophagic cell death in ovarian cancer cells. Furthermore, human whole genome expression profile microarrays and massage pathway analysis revealed that during early stages of autophagy in ovarian cancer cells with highly expressing Lewis y antigen, PI3K/Akt-mTOR activity was reduced, in contrast, the PI3K/Akt-mTOR signaling pathway was activated as the length of amino acid deprivation increased, which inhibited eIF4G2 expression, further decreased the transcription of autophagy-related genes, suppressed autophagic cell death. we also elaborated that co-regulates protein degradation in cells via the ubiquitin-proteasome system and the autophagy-lysosome pathway. These findings suggested that the modification of CD147 by Lewis y antigen enhanced the survival ability by promoting basic autophagy activity and restraining autophagic cell death in ovarian cancer , thus playing an important role in ovarian cancer malignant progression.
Collapse
Affiliation(s)
- Zhenhua Hu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Mingbo Cai
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
| | - Lu Deng
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
| | - Liancheng Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
| | - Jian Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
| | - Mingzi Tan
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
| | - Juanjuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, 110004, China
| |
Collapse
|
11
|
Joshi S, Kumar S, Ponnusamy MP, Batra SK. Hypoxia-induced oxidative stress promotes MUC4 degradation via autophagy to enhance pancreatic cancer cells survival. Oncogene 2016; 35:5882-5892. [PMID: 27109098 DOI: 10.1038/onc.2016.119] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 12/06/2015] [Accepted: 01/04/2016] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer (PC) and associated pre-neoplastic lesions have been reported to be hypoxic, primarily due to hypovascular nature of PC. Though the presence of hypoxia under cancerous condition has been associated with the overexpression of oncogenic proteins (MUC1), multiple emerging reports have also indicated the growth inhibitory effects of hypoxia. In spite of being recognized as the top-most differentially expressed and established oncogenic protein in PC, MUC4 regulation in terms of micro-environmental stress has not been determined. Herein, for the first time, we are reporting that MUC4 protein stability is drastically affected in PC, under hypoxic condition in a hypoxia inducible factor 1α (HIF-1α)-independent manner. Mechanistically, we have demonstrated that hypoxia-mediated induction of reactive oxygen species (ROS) promotes autophagy by inhibiting pAkt/mTORC1 pathway, one of the central regulators of autophagy. Immunohistofluorescence analyses revealed significant negative correlation (P-value=0.017) between 8-hydroxy guanosine (8-OHG) and MUC4 in primary pancreatic tumors (n=25). Moreover, we found pronounced colocalization between MUC4 and LAMP1/LC3 (microtubule-associated protein 1A/1B-light chain 3) in PC tissues and also observed their negative relationship in their expression pattern, suggesting that areas with high autophagy rate had less MUC4 expression. We also found that hypoxia and ROS have negative impact on overall cell growth and viability, which was partially, though significantly (P<0.05), rescued in the presence of MUC4. Altogether, hypoxia-mediated oxidative stress induces autophagy in PC, leading to the MUC4 degradation to enhance survival, possibly by offering required metabolites to stressed cells.
Collapse
Affiliation(s)
- S Joshi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - M P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
12
|
Abstract
BECN1/Beclin 1 is regarded as a critical component in the class III phosphatidylinositol 3-kinase (PtdIns3K) complex to trigger autophagy in mammalian cells. Despite its significant role in a number of cellular and physiological processes, the exact function of BECN1 in autophagy remains controversial. Here we created a BECN1 knockout human cell line using the TALEN technique. Surprisingly, the complete loss of BECN1 had little effect on LC3 (MAP1LC3B/LC3B) lipidation, and LC3B puncta resembling autophagosomes by fluorescence microscopy were still evident albeit significantly smaller than those in the wild-type cells. Electron microscopy (EM) analysis revealed that BECN1 deficiency led to malformed autophagosome-like structures containing multiple layers of membranes under amino acid starvation. We further confirmed that the PtdIns3K complex activity and autophagy flux were disrupted in BECN1−/− cells. Our results demonstrate the essential role of BECN1 in the functional formation of autophagosomes, but not in LC3B lipidation.
Collapse
Key Words
- ATG, autophagy related
- BCL2L1/Bcl-xL, BCL2-like 1
- BECN1
- BECN1, Beclin 1, autophagy related
- BECN1P1/BECN2, Beclin 1, autophagy related, pseudogene 1
- BafA1, bafilomycin A1
- EM, electron microscopy
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GFP, green fluorescent protein
- KO, knockout
- LC3
- MAP1LC3-I/LC3-I, soluble, proteolytically processed microtubule-associated protein 1 light chain 3
- MAP1LC3-II/LC3-II, proteolytically processed and lipid-modified microtubule-associated protein 1 light chain 3
- MAP1LC3/LC3, microtubule-associated protein 1 light chain 3
- MAP1LC3B/LC3B, microtubule-associated protein 1 light chain 3 β
- PIK3C3/VPS34, phosphatidylinositol 3-kinase, catalytic subunit type 3
- PIK3R4/VPS15, phosphoinositide-3-kinase, regulatory subunit 4
- PtdIns3K
- PtdIns3K, phosphatidylinositol 3-kinase
- SQSTM1/p62, sequestosome 1
- TALEN, transcription activator-like effector nuclease
- TUBB, tubulin, β class I
- UVRAG, UV radiation resistance associated
- ZFYVE1/DFCP1, zinc finger, FYVE domain containing 1.
- autophagosome
- autophagy
Collapse
Affiliation(s)
- Ruina He
- a Biodynamic Optical Imaging Center (BIOPIC); Peking-Tsinghua Center for Life Sciences; State Key Laboratory of Protein and Plant Gene Research; School of Life Sciences; Peking University ; Beijing , China
| | | | | | | | | |
Collapse
|
13
|
Galluzzi L, Pietrocola F, Bravo-San Pedro JM, Amaravadi RK, Baehrecke EH, Cecconi F, Codogno P, Debnath J, Gewirtz DA, Karantza V, Kimmelman A, Kumar S, Levine B, Maiuri MC, Martin SJ, Penninger J, Piacentini M, Rubinsztein DC, Simon HU, Simonsen A, Thorburn AM, Velasco G, Ryan KM, Kroemer G. Autophagy in malignant transformation and cancer progression. EMBO J 2015; 34:856-80. [PMID: 25712477 PMCID: PMC4388596 DOI: 10.15252/embj.201490784] [Citation(s) in RCA: 916] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/15/2022] Open
Abstract
Autophagy plays a key role in the maintenance of cellular homeostasis. In healthy cells, such a homeostatic activity constitutes a robust barrier against malignant transformation. Accordingly, many oncoproteins inhibit, and several oncosuppressor proteins promote, autophagy. Moreover, autophagy is required for optimal anticancer immunosurveillance. In neoplastic cells, however, autophagic responses constitute a means to cope with intracellular and environmental stress, thus favoring tumor progression. This implies that at least in some cases, oncogenesis proceeds along with a temporary inhibition of autophagy or a gain of molecular functions that antagonize its oncosuppressive activity. Here, we discuss the differential impact of autophagy on distinct phases of tumorigenesis and the implications of this concept for the use of autophagy modulators in cancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Federico Pietrocola
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France
| | - José Manuel Bravo-San Pedro
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France
| | - Ravi K Amaravadi
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Francesco Cecconi
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark IRCCS Fondazione Santa Lucia and Department of Biology University of Rome Tor Vergata, Rome, Italy
| | - Patrice Codogno
- Université Paris Descartes Sorbonne Paris Cité, Paris, France Institut Necker Enfants-Malades (INEM), Paris, France INSERM U1151, Paris, France CNRS UMR8253, Paris, France
| | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - David A Gewirtz
- Department of Pharmacology, Toxicology and Medicine, Virginia Commonwealth University, Richmond Virginia, VA, USA
| | | | - Alec Kimmelman
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maria Chiara Maiuri
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Gustave Roussy Cancer Campus, Villejuif, France
| | - Seamus J Martin
- Department of Genetics, Trinity College, The Smurfit Institute, Dublin, Ireland
| | - Josef Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Mauro Piacentini
- Department of Biology, University of Rome Tor Vergata, Rome, Italy National Institute for Infectious Diseases IRCCS 'Lazzaro Spallanzani', Rome, Italy
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Andrew M Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology I, School of Biology, Complutense University of Madrid, Madrid, Spain Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Guido Kroemer
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France INSERM U1138, Paris, France Université Paris Descartes Sorbonne Paris Cité, Paris, France Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
14
|
Li RF, Chen G, Ren JG, Zhang W, Wu ZX, Liu B, Zhao Y, Zhao YF. The adaptor protein p62 is involved in RANKL-induced autophagy and osteoclastogenesis. J Histochem Cytochem 2014; 62:879-88. [PMID: 25163928 DOI: 10.1369/0022155414551367] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Previous studies have implicated autophagy in osteoclast differentiation. The aim of this study was to investigate the potential role of p62, a characterized adaptor protein for autophagy, in RANKL-induced osteoclastogenesis. Real-time quantitative PCR and western blot analyses were used to evaluate the expression levels of autophagy-related markers during RANKL-induced osteoclastogenesis in mouse macrophage-like RAW264.7 cells. Meanwhile, the potential relationship between p62/LC3 localization and F-actin ring formation was tested using double-labeling immunofluorescence. Then, the expression of p62 in RAW264.7 cells was knocked down using small-interfering RNA (siRNA), followed by detecting its influence on RANKL-induced autophagy activation, osteoclast differentiation, and F-actin ring formation. The data showed that several key autophagy-related markers including p62 were significantly altered during RANKL-induced osteoclast differentiation. In addition, the expression and localization of p62 showed negative correlation with LC3 accumulation and F-actin ring formation, as demonstrated by western blot and immunofluorescence analyses, respectively. Importantly, the knockdown of p62 obviously attenuated RANKL-induced expression of autophagy- and osteoclastogenesis-related genes, formation of TRAP-positive multinuclear cells, accumulation of LC3, as well as formation of F-actin ring. Our study indicates that p62 may play essential roles in RANKL-induced autophagy and osteoclastogenesis, which may help to develop a novel therapeutic strategy against osteoclastogenesis-related diseases.
Collapse
Affiliation(s)
- Rui-Fang Li
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| | - Jian-Gang Ren
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| | - Wei Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| | - Zhong-Xing Wu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| | - Yi Zhao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| | - Yi-Fang Zhao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology (RFL, GC, JGR, WZ, ZXW, BL, YZ, YFZ) Wuhan University, Wuhan, ChinaDepartment of Oral and Maxillofacial Surgery, School & Hospital of Stomatology (GC, ZXW, BL, YFZ) Wuhan University, Wuhan, ChinaDepartment of Prosthodontics, School & Hospital of Stomatology (YZ) Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Expression and clinical significance of the autophagy proteins BECLIN 1 and LC3 in ovarian cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:462658. [PMID: 25136588 PMCID: PMC4127242 DOI: 10.1155/2014/462658] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 06/27/2014] [Indexed: 12/13/2022]
Abstract
Autophagy is dysregulated in cancer and might be involved in ovarian carcinogenesis. BECLIN-1, a protein that interacts with either BCL-2 or PI3k class III, plays a critical role in the regulation of both autophagy and cell death. Induction of autophagy is associated with the presence of vacuoles characteristically labelled with the protein LC3. We have studied the biological and clinical significance of BECLIN 1 and LC3 in ovary tumours of different histological types. The positive expression of BECLIN 1 was well correlated with the presence of LC3-positive autophagic vacuoles and was inversely correlated with the expression of BCL-2. The latter inhibits the autophagy function of BECLIN 1. We found that type I tumours, which are less aggressive than type II, were more frequently expressing high level of BECLIN 1. Of note, tumours of histologic grade III expressed low level of BECLIN 1. Consistently, high level of expression of BECLIN 1 and LC3 in tumours is well correlated with the overall survival of the patients. The present data are compatible with the hypotheses that a low level of autophagy favours cancer progression and that ovary cancer with upregulated autophagy has a less aggressive behaviour and is more responsive to chemotherapy.
Collapse
|
16
|
Li RF, Chen G, Zhao Y, Zhao YF, Liu B. Increased expression of autophagy-related proteins in keratocystic odontogenic tumours: its possible association with growth potential. Br J Oral Maxillofac Surg 2014; 52:551-6. [PMID: 24703774 DOI: 10.1016/j.bjoms.2014.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 03/07/2014] [Indexed: 01/17/2023]
Abstract
The aim of this study was to evaluate the activation status of autophagy in keratocystic odontogenic tumours (KCOT), and to investigate its possible association with growth potential. We detected the expression of some key autophagy-related proteins in clinical samples of KCOT and radicular cysts and compared then by real-time quantitative polymerase chain reaction (qPCR) and immunohistochemical analysis, respectively. The correlation between the autophagy-related proteins tested, and with cell antiapoptotic (Bcl-2) or proliferative (Ki-67) markers in KCOT was explored using Spearman's rank correlation, followed by cluster analysis. The results showed that both the expression of mRNA and the immunoreactivity of the autophagy-related proteins tested were considerably increased in samples of KCOT compared with those in samples of radicular cysts. The correlation analyses showed that the immunostains of autophagy-related proteins in samples of KCOT correlated closely with each other. The immunostains of these autophagy-related proteins also correlated closely with the immunostains of Bcl-2 and Ki-67 in KCOT. More importantly, double-labelling immunofluorescence analyses also showed that the distribution of autophagic and proliferative markers was partially synchronous in samples from KCOT. We have, to our knowledge for the first time, implicated the activation of autophagy in KCOT, and showed its possible association with growth potential.
Collapse
Affiliation(s)
- Rui-Fang Li
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, China
| | - Gang Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Yi Zhao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, China; Department of Prosthodontics, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Yi-Fang Zhao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
17
|
Chondrogianni N, Sakellari M, Lefaki M, Papaevgeniou N, Gonos ES. Proteasome activation delays aging in vitro and in vivo. Free Radic Biol Med 2014; 71:303-320. [PMID: 24681338 DOI: 10.1016/j.freeradbiomed.2014.03.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 02/02/2023]
Abstract
Aging is a natural biological process that is characterized by a progressive accumulation of macromolecular damage. In the proteome, aging is accompanied by decreased protein homeostasis and function of the major cellular proteolytic systems, leading to the accumulation of unfolded, misfolded, or aggregated proteins. In particular, the proteasome is responsible for the removal of normal as well as damaged or misfolded proteins. Extensive work during the past several years has clearly demonstrated that proteasome activation by either genetic means or use of compounds significantly retards aging. Importantly, this represents a common feature across evolution, thereby suggesting proteasome activation to be an evolutionarily conserved mechanism of aging and longevity regulation. This review article reports on the means of function of these proteasome activators and how they regulate aging in various species.
Collapse
Affiliation(s)
- Niki Chondrogianni
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry, and Biotechnology, 116 35 Athens, Greece.
| | - Marianthi Sakellari
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry, and Biotechnology, 116 35 Athens, Greece; Örebro University Medical School, Örebro, Sweden
| | - Maria Lefaki
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry, and Biotechnology, 116 35 Athens, Greece
| | - Nikoletta Papaevgeniou
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry, and Biotechnology, 116 35 Athens, Greece
| | - Efstathios S Gonos
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry, and Biotechnology, 116 35 Athens, Greece; Örebro University Medical School, Örebro, Sweden
| |
Collapse
|
18
|
Manipulation of autophagy in cancer cells: an innovative strategy to fight drug resistance. Future Med Chem 2013; 5:1009-21. [PMID: 23734684 DOI: 10.4155/fmc.13.85] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a catabolic process activated by stress conditions and nutrient deprivation, to which it reacts by promoting the degradation of damaged organelles and misfolded/aggregated proteins, as well as generating new energetic pools. Paradoxically, in cancer cells, which signal the dangerous microenvironment occurring during clinical therapies, autophagy could promote their proliferation and sustain drug resistance. Special attention is given to autophagy manipulation in order to counteract drug resistance of cancer cells. This article describes the basic properties of autophagy and focuses on the strategies of manipulating it.
Collapse
|
19
|
Kyrychenko VO, Nagibin VS, Tumanovska LV, Pashevin DO, Gurianova VL, Moibenko AA, Dosenko VE, Klionsky DJ. Knockdown of PSMB7 induces autophagy in cardiomyocyte cultures: possible role in endoplasmic reticulum stress. Pathobiology 2013; 81:8-14. [PMID: 23969338 DOI: 10.1159/000350704] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 03/11/2013] [Indexed: 11/19/2022] Open
Abstract
Proteasomal and autophagic pathways of protein degradation are two essential, endoplasmic reticulum (ER)-associated proteolytic systems involved in the ER stress response. The functional interaction between them has been shown by proteasome pharmacological inhibition. However, little data have been found concerning autophagy induction using an RNA interference approach due to the multisubunit composition of proteolytic systems. We suggested that silencing of single proteasome subunits can induce massive autophagy. Psmb7-specific small interference RNA added to isolated cardiomyocytes significantly affected mRNA expression of essential ER stress marker proteins, including DDIT3/CHOP and HSPA5/GRP78. mRNA expression of the key autophagy regulator MTOR was also increased. These findings were confirmed by single-cell reverse transcription real-time PCR on individual monodansylcadaverine (MDC)-labeled cardiomyocytes. RNA interference that decreased the levels of non-catalytic PSMB7 subunits of the proteasome had no influence on chymotrypsin- and trypsin-like activities, but significantly decreased peptidyl-glutamyl peptide-hydrolyzing activity. Immunohistochemical analysis showed increased levels of LC3-marked vacuoles in the cytoplasm of Psmb7-knockdown cells, and MDC staining showed significantly increased numbers of neonatal cardiomyocytes with autophagic vacuoles. After anoxia-reoxygenation, the number of cells with signs of autophagy after Psmb7 gene silencing was higher. Our results indicate that Psmb7 knockdown induces ER stress and autophagy in cardiomyocytes, which may be a useful approach to activate specific autophagy.
Collapse
Affiliation(s)
- Victoria O Kyrychenko
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Walker DM, Mahfooz N, Kemme KA, Patel VC, Spangler M, Drew ME. Plasmodium falciparum erythrocytic stage parasites require the putative autophagy protein PfAtg7 for normal growth. PLoS One 2013; 8:e67047. [PMID: 23825614 PMCID: PMC3692556 DOI: 10.1371/journal.pone.0067047] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 05/13/2013] [Indexed: 11/19/2022] Open
Abstract
Analysis of the Plasmodium falciparum genome reveals a limited number of putative autophagy genes, specifically the four genes involved in ATG8 lipidation, an essential step in formation of autophagosomes. In yeast, Atg8 lipidation requires the E1-type ligase Atg7, an E2-type ligase Atg3, and a cysteine protease Atg4. These four putative P. falciparum ATG (PfATG) genes are transcribed during the parasite's erythrocytic stages. PfAtg7 has relatively low identity and similarity to yeast Atg7 (14.7% and 32.2%, respectively), due primarily to long insertions typical of P. falciparum. Excluding the insertions the identity and similarity are higher (38.0% and 70.8%, respectively). This and the fact that key residues are conserved, including the catalytic cysteine and ATP binding domain, we hypothesize that PfAtg7 is the activating enzyme of PfAtg8. To assess the role of PfAtg7 we have generated two transgenic parasite lines. In one, the PfATG7 locus was modified to introduce a C-terminal hemagglutinin tag. Western blotting reveals two distinct protein species, one migrating near the predicted 150 kDa and one at approximately 65 kDa. The second transgenic line introduces an inducible degradation domain into the PfATG7 locus, allowing us to rapidly attenuate PfAtg7 protein levels. Corresponding species are also observed in this parasite line at approximately 200 kDa and 100 kDa. Upon PfATG7 attenuation parasites exhibit a slow growth phenotype indicating the essentiality of this putative enzyme for normal growth.
Collapse
Affiliation(s)
- Dawn M. Walker
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Najmus Mahfooz
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Katherine A. Kemme
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Viral C. Patel
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Maribeth Spangler
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Mark E. Drew
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Division of Medicinal Chemistry, The Ohio State University College of Pharmacy, Columbus, Ohio, United States of America
| |
Collapse
|