1
|
ALKhemeiri N, Eljack S, Saber-Ayad MM. Perspectives of Targeting Autophagy as an Adjuvant to Anti-PD-1/PD-L1 Therapy for Colorectal Cancer Treatment. Cells 2025; 14:745. [PMID: 40422248 DOI: 10.3390/cells14100745] [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: 02/13/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world, with increasing incidence and mortality rates. Standard conventional treatments for CRC are surgery, chemotherapy, and radiotherapy. Recently, immunotherapy has been introduced as a promising alternative to CRC treatment that utilizes patients' immune system to combat cancer cells. The beneficial effect of immune checkpoint inhibitors, specifically anti-PD-1/ PD-L1, has been ascribed to the abundance of DNA replication errors that result in the formation of neoantigens. Such neoantigens serve as distinct flags that amplify the immune response when checkpoint inhibitors (ICIs) are administered. DNA replication errors in CRC patients are expressed as two statuses: the first is the deficient mismatch repair (MSI-H/dMMR) with a higher overall immune response and survival rate than the second status of patients with proficient mismatch repair (MSS/pMMR). There is a limitation to using anti-PD-1/PD-L1 as it is only confined to MSI-H/dMMR, where there is an abundance of T-cell inhibitory ligands (PD-L1). This calls for investigating new therapeutic interventions to widen the scope of ICIs' role in the treatment of CRC. Autophagy modulation provides a good example. Autophagy is a cellular process that plays a crucial role in maintaining cellular homeostasis and has been studied for its impact on tumor development, progression, and response to treatment. In this review, we aim to highlight autophagy as a potential determinant in tumor immune response and to study the impact of autophagy on the tumor immune microenvironment. Moreover, we aim to investigate the value of a combination of anti-PD-1/PD-L1 agents with autophagy modulators as an adjuvant therapeutic approach for CRC treatment.
Collapse
Affiliation(s)
- Nasrah ALKhemeiri
- College of Graduate Studies, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Sahar Eljack
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Maha Mohamed Saber-Ayad
- College of Graduate Studies, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Department of Pharmacology, Faculty of Medicine, Cairo University, Cairo 12211, Egypt
| |
Collapse
|
2
|
Liu J, Zhang F, Yan Z, Guo Z, Lu Y, Yao B, Li Y, Lv W. Effects of prolonged NaHCO 3 exposure on the serum immune function, antioxidant capacity, intestinal tight junctions, microbiota, mitochondria, and autophagy in crucian carp (Carassius auratus). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117571. [PMID: 39708453 DOI: 10.1016/j.ecoenv.2024.117571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
This study investigated the effects of long-term NaHCO3 stress on serum immunity, antioxidant capacity, intestinal tight junction, intestinal microbiota, mitochondrial function and autophagy in crucian carp. A total of 240 fish (31.19 ± 1.03 g) were randomly allocated to two groups and treated with 0 mmol/L (CK) and 50 mmol/L NaHCO3 (CA) respectively, to simulate the carbonate alkaline water environment. All of the experimental fish were cultured for 8 weeks. The results showed that compared to the control group, NaHCO3 stress significantly decreased the levels of the serum immunity indices (AKP, ACP, C3, C4, IgM, LZM) and the antioxidant capacity indices (CAT, GSH-PX, SOD, T-AOC), while markedly increasing the content of MDA. Additionally, NaHCO3 influenced the mRNA expression of HSP90, Nrf2, Keap1, and HO-1. Compared to the control group, the levels of ZO-1, Claudin-2, Occludin-a, and Occludin-b mRNA significantly decreased in the NaHCO3 stress group. The levels of ATG5, ATG7, and Beclin1 mRNA and protein were significantly increased along with the levels of LC3b mRNA and the ratio of protein LC3 II /LC3 I. Compared to the control group, intestinal mitochondria in the NaHCO3 stress group were visibly swollen and largely broken, with reductions in ridges and a large proportion of the area dissolved in the matrix. The mitochondrial membrane potential and the activities of ATPase were significantly decreased, leading to mitochondria dysfunction. In addition, 3147 differentially expressed genes were identified from transcriptome sequencing, among which several genes related to mitochondria and autophagy were significantly enriched. Compared to the control group, the NaHCO3 stress decreased the ACE index and increased the abundance of Proteobacteria while decreased the abundance of Actinobacteria and Firmicutes. In conclusion, NaHCO3 induced oxidative damage, microbiota alterations, mitochondria dysfunction, and autophagy in the intestines of crucian carp. The results of this study have characterized the molecular mechanisms of intestinal injury in crucian carp caused by NaHCO3 stress, and thus provide empirical support for aquaculture in saline-alkali waters.
Collapse
Affiliation(s)
- Jia Liu
- College of Animal Science and Technology/College of Animal Medicine, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Faye Zhang
- Georgia Institute of Technology, Atlanta, USA
| | - Zihao Yan
- College of Animal Science and Technology/College of Animal Medicine, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Zhengyao Guo
- College of Animal Science and Technology/College of Animal Medicine, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Yuqian Lu
- College of Animal Science and Technology/College of Animal Medicine, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Baolan Yao
- College of Animal Science and Technology/College of Animal Medicine, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China
| | - Yuehong Li
- College of Animal Science and Technology/College of Animal Medicine, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China.
| | - Wenfa Lv
- College of Animal Science and Technology/College of Animal Medicine, Jilin Agricultural University, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
3
|
Crotts MS, Jacobs JC, Baer RW, Cox JL. Doramectin Induces Apoptosis in B16 Melanoma Cells. Anticancer Agents Med Chem 2025; 25:244-256. [PMID: 39411968 DOI: 10.2174/0118715206325844240909144543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 03/25/2025]
Abstract
INTRODUCTION/OBJECTIVE Metastatic melanoma resists current pharmacological regimens that act through apoptosis. This indicates that therapies acting via non-apoptotic cell-death pathways could be pursued. Doramectin has shown promising results in another cancer of neural crest origin, neuroblastoma, through the inhibition of growth via autophagy. Our research hypothesis is that doramectin induces autophagy in B16F10 melanoma cells. METHODS Cells were treated with doramectin (15 uM) or a combination of both doramectin and a cell-death inhibitor, compared to untreated control cells (media), and then analyzed with MTT analysis. Likewise, MDC analysis was completed to detect autophagy involvement with doramectin treatment. Flow cytometry and TUNEL Assay were conducted to observe cell death-related effects. RESULTS MTT analysis of doramectin-treated cells displayed a decrease in cell growth compared to control. Apoptotic morphology was prominent in melanoma cells treated with doramectin. Increased autophagy was not detected by fluorometric microscopic analysis. Flow cytometry analysis of doramectin-treated cells showed apoptosis as a major mode of cell death with some necrosis. CONCLUSION Doramectin induces a novel cell-death mechanism in melanoma compared to other forms of cancer and should be studied as an effective anti-cancer agent for melanoma treatment.
Collapse
Affiliation(s)
- Megan S Crotts
- Department of Biochemistry, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
| | - Jena C Jacobs
- Department of Biochemistry, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
| | - Robert W Baer
- Department of Biochemistry, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
| | - James L Cox
- Department of Biochemistry, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, Missouri, USA
| |
Collapse
|
4
|
Kovale L, Singh MK, Kim J, Ha J. Role of Autophagy and AMPK in Cancer Stem Cells: Therapeutic Opportunities and Obstacles in Cancer. Int J Mol Sci 2024; 25:8647. [PMID: 39201332 PMCID: PMC11354724 DOI: 10.3390/ijms25168647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer stem cells represent a resilient subset within the tumor microenvironment capable of differentiation, regeneration, and resistance to chemotherapeutic agents, often using dormancy as a shield. Their unique properties, including drug resistance and metastatic potential, pose challenges for effective targeting. These cells exploit certain metabolic processes for their maintenance and survival. One of these processes is autophagy, which generally helps in energy homeostasis but when hijacked by CSCs can help maintain their stemness. Thus, it is often referred as an Achilles heel in CSCs, as certain cancers tend to depend on autophagy for survival. Autophagy, while crucial for maintaining stemness in cancer stem cells (CSCs), can also serve as a vulnerability in certain contexts, making it a complex target for therapy. Regulators of autophagy like AMPK (5' adenosine monophosphate-activated protein kinase) also play a crucial role in maintaining CSCs stemness by helping CSCs in metabolic reprogramming in harsh environments. The purpose of this review is to elucidate the interplay between autophagy and AMPK in CSCs, highlighting the challenges in targeting autophagy and discussing therapeutic strategies to overcome these limitations. This review focuses on previous research on autophagy and its regulators in cancer biology, particularly in CSCs, addresses the remaining unanswered questions, and potential targets for therapy are also brought to attention.
Collapse
Affiliation(s)
- Lochana Kovale
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| | - Joungmok Kim
- Department of Oral Biochemistry and Molecular Biology, College of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (L.K.); (M.K.S.)
| |
Collapse
|
5
|
Ahmadi-Dehlaghi F, Mohammadi P, Valipour E, Pournaghi P, Kiani S, Mansouri K. Autophagy: A challengeable paradox in cancer treatment. Cancer Med 2023. [PMID: 36760166 DOI: 10.1002/cam4.5577] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/14/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE Autophagy is an intracellular degradation pathway conserved in all eukaryotes from yeast to humans. This process plays a quality-control role by destroying harmful cellular components under normal conditions, maintaining cell survival, and establishing cellular adaptation under stressful conditions. Hence, there are various studies indicating dysfunctional autophagy as a factor involved in the development and progression of various human diseases, including cancer. In addition, the importance of autophagy in the development of cancer has been highlighted by paradoxical roles, as a cytoprotective and cytotoxic mechanism. Despite extensive research in the field of cancer, there are many questions and challenges about the roles and effects suggested for autophagy in cancer treatment. The aim of this study was to provide an overview of the paradoxical roles of autophagy in different tumors and related cancer treatment options. METHODS In this study, to find articles, a search was made in PubMed and Google scholar databases with the keywords Autophagy, Autophagy in Cancer Management, and Drug Design. RESULTS According to the investigation, some studies suggest that several advanced cancers are dependent on autophagy for cell survival, so when cancer cells are exposed to therapy, autophagy is induced and suppresses the anti-cancer effects of therapeutic agents and also results in cell resistance. However, enhanced autophagy from using anti-cancer drugs causes autophagy-mediated cell death in several cancers. Because autophagy also plays roles in both tumor suppression and promotion further research is needed to determine the precise mechanism of this process in cancer treatment. CONCLUSION We concluded in this article, autophagy manipulation may either promote or hinder the growth and development of cancer according to the origin of the cancer cells, the type of cancer, and the behavior of the cancer cells exposed to treatment. Thus, before starting treatment it is necessary to determine the basal levels of autophagy in various cancers.
Collapse
Affiliation(s)
- Farnaz Ahmadi-Dehlaghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Biology, Payame Noor University, Tehran, Iran
| | - Parisa Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elahe Valipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sarah Kiani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
6
|
Al-Bari AA. Inhibition of autolysosomes by repurposing drugs as a promising therapeutic strategy for the treatment of cancers. ALL LIFE 2022; 15:568-601. [DOI: 10.1080/26895293.2022.2078894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/02/2022] [Indexed: 10/18/2022] Open
Affiliation(s)
- Abdul Alim Al-Bari
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
7
|
Kao CH, Su TY, Huang WS, Lu XY, Jane WN, Huang CY, Huang HH, Wang WJ. TFEB- and TFE3-dependent autophagy activation supports cancer proliferation in the absence of centrosomes. Autophagy 2022; 18:2830-2850. [PMID: 35316161 PMCID: PMC9673955 DOI: 10.1080/15548627.2022.2051880] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Centrosome amplification is a phenomenon frequently observed in human cancers, so centrosome depletion has been proposed as a therapeutic strategy. However, despite being afflicted with a lack of centrosomes, many cancer cells can still proliferate, implying there are impediments to adopting centrosome depletion as a treatment strategy. Here, we show that TFEB- and TFE3-dependent autophagy activation contributes to acentrosomal cancer proliferation. Our biochemical analyses uncover that both TFEB and TFE3 are novel PLK4 (polo like kinase 4) substrates. Centrosome depletion inactivates PLK4, resulting in TFEB and TFE3 dephosphorylation and subsequent promotion of TFEB and TFE3 nuclear translocation and transcriptional activation of autophagy- and lysosome-related genes. A combination of centrosome depletion and inhibition of the TFEB-TFE3 autophagy-lysosome pathway induced strongly anti-proliferative effects in cancer cells. Thus, our findings point to a new strategy for combating cancer.Abbreviations: AdCre: adenoviral Cre recombinase; AdLuc: adenoviral luciferase; ATG5: autophagy related 5; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; DKO: double knockout; GFP: green fluorescent protein; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LAMP2: lysosomal associated membrane protein 2; LTR: LysoTracker Red; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MITF: melanocyte inducing transcription factor; PLK4: polo like kinase 4; RFP: red fluorescent protein; SASS6: SAS-6 centriolar assembly protein; STIL: STIL centriolar assembly protein; TFEB: transcription factor EB; TFEBΔNLS: TFEB lacking a nuclear localization signal; TFE3: transcription factor binding to IGHM enhancer 3; TP53/p53: tumor protein p53.
Collapse
Affiliation(s)
- Chien-Han Kao
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, TaipeiTaiwan
| | - Ting-Yu Su
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, TaipeiTaiwan
| | - Wei-Syun Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, TaipeiTaiwan
| | - Xin-Ying Lu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, TaipeiTaiwan
| | - Wann-Neng Jane
- Institute of Plant and Microbial Biology, Academia Sinica, Taiwan
| | - Chien-Yung Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, TaipeiTaiwan
| | - Hung-Hsiang Huang
- Division of Urology, Department of Surgery, Far Eastern Memorial Hospital, New Taipei CityTaiwan
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, TaipeiTaiwan
- CONTACT Won-Jing Wang Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, TaipeiTaiwan
| |
Collapse
|
8
|
Mainz L, Sarhan MAFE, Roth S, Sauer U, Kalogirou C, Eckstein M, Gerhard-Hartmann E, Seibert HD, Voelker HU, Geppert C, Rosenwald A, Eilers M, Schulze A, Diefenbacher M, Rosenfeldt MT. Acute systemic knockdown of Atg7 is lethal and causes pancreatic destruction in shRNA transgenic mice. Autophagy 2022; 18:2880-2893. [PMID: 35343375 PMCID: PMC9673934 DOI: 10.1080/15548627.2022.2052588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The notion that macroautophagy/autophagy is a potentially attractive therapeutic target for a variety of diseases, including cancer, largely stems from pre-clinical mouse studies. Most of these examine the effects of irreversible and organ confined autophagy deletion using site specific Cre-loxP recombination of the essential autophagy regulating genes Atg7 or Atg5. Model systems with the ability to impair autophagy systemically and reversibly at all disease stages would allow a more realistic approach to evaluate the consequences of authophagy inhibition as a therapeutic concept and its potential side effects. Here, we present shRNA transgenic mice that via doxycycline (DOX) regulable expression of a highly efficient miR30-E-based shRNA enabled knockdown of Atg7 simultaneously in the majority of organs, with the brain and spleen being noteable exceptions. Induced animals deteriorated rapidly and experienced profound destruction of the exocrine pancreas, severe hypoglycemia and depletion of hepatic glycogen storages. Cessation of DOX application restored apparent health, glucose homeostasis and pancreatic integrity. In a similar Atg5 knockdown model we neither observed loss of pancreatic integrity nor diminished survival after DOX treatment, but identified histological changes consistent with steatohepatitis and hepatic fibrosis in the recovery period after termination of DOX. Regulable Atg7-shRNA mice are valuable tools that will enable further studies on the role of autophagy impairment at various disease stages and thereby help to evaluate the consequences of acute autophagy inhibition as a therapeutic concept.Abbreviations: ACTB: actin, beta; AMY: amylase complex; ATG4B: autophagy related 4B, cysteine peptidase; ATG5: autophagy related 5; ATG7: autophagy related 7; Cag: CMV early enhancer/chicken ACTB promoter; Col1a1: collagen, type I, alpha 1; Cre: cre recombinase; DOX: doxycycline; GCG: glucagon; GFP: green fluorescent protein; INS: insulin; LC3: microtubule-associated protein 1 light chain 3; miR30-E: optimized microRNA backbone; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; PNLIP: pancreatic lipase; rtTA: reverse tetracycline transactivator protein; SQSTM1/p62: sequestome 1; TRE: tetracycline responsive element.
Collapse
Affiliation(s)
- Laura Mainz
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Mohamed A. F. E. Sarhan
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Sabine Roth
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Ursula Sauer
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Charis Kalogirou
- Department of Urology and Pediatric Urology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elena Gerhard-Hartmann
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Helen-Desiree Seibert
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Hans-Ulrich Voelker
- Department of Pathology, Leopoldina Medizinisches Versorgungszentrum, Schweinfurt, Germany
| | - Carol Geppert
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Martin Eilers
- Biocenter, Department of Biochemistry and Molecular Biology, Julius-Maximilians-University of Würzburg, Germany
| | - Almut Schulze
- Division of Metabolism and Microenvironment, Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), Germany
| | - Markus Diefenbacher
- Biocenter, Department of Biochemistry and Molecular Biology, Julius-Maximilians-University of Würzburg, Germany
| | - Mathias T. Rosenfeldt
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany,Comprehensive Cancer Center Mainfranke, Julius-Maximilians-University of Würzburg, Würzburg, Germany,CONTACT Mathias T. Rosenfeldt Institute of Pathology – University of Würzburg, Josef-Schneider-Str. 2,97080Würzburg, Germany
| |
Collapse
|
9
|
Mainz L, Sarhan MAFE, Roth S, Sauer U, Maurus K, Hartmann EM, Seibert HD, Rosenwald A, Diefenbacher ME, Rosenfeldt MT. Autophagy Blockage Reduces the Incidence of Pancreatic Ductal Adenocarcinoma in the Context of Mutant Trp53. Front Cell Dev Biol 2022; 10:785252. [PMID: 35372352 PMCID: PMC8965752 DOI: 10.3389/fcell.2022.785252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) is a homeostatic process that preserves cellular integrity. In mice, autophagy regulates pancreatic ductal adenocarcinoma (PDAC) development in a manner dependent on the status of the tumor suppressor gene Trp53. Studies published so far have investigated the impact of autophagy blockage in tumors arising from Trp53-hemizygous or -homozygous tissue. In contrast, in human PDACs the tumor suppressor gene TP53 is mutated rather than allelically lost, and TP53 mutants retain pathobiological functions that differ from complete allelic loss. In order to better represent the patient situation, we have investigated PDAC development in a well-characterized genetically engineered mouse model (GEMM) of PDAC with mutant Trp53 (Trp53R172H) and deletion of the essential autophagy gene Atg7. Autophagy blockage reduced PDAC incidence but had no impact on survival time in the subset of animals that formed a tumor. In the absence of Atg7, non-tumor-bearing mice reached a similar age as animals with malignant disease. However, the architecture of autophagy-deficient, tumor-free pancreata was effaced, normal acinar tissue was largely replaced with low-grade pancreatic intraepithelial neoplasias (PanINs) and insulin expressing islet β-cells were reduced. Our data add further complexity to the interplay between Atg7 inhibition and Trp53 status in tumorigenesis.
Collapse
Affiliation(s)
- Laura Mainz
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Salk Institute for Biological Studies, San Diego, CA, United States
| | - Mohamed A. F. E. Sarhan
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Sabine Roth
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Ursula Sauer
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Katja Maurus
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Elena M. Hartmann
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Helen-Desiree Seibert
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Markus E. Diefenbacher
- Biocenter, Department of Biochemistry and Molecular Biology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Mathias T. Rosenfeldt
- Institute of Pathology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
- *Correspondence: Mathias T. Rosenfeldt,
| |
Collapse
|
10
|
Diehl V, Huber LS, Trebicka J, Wygrecka M, Iozzo RV, Schaefer L. The Role of Decorin and Biglycan Signaling in Tumorigenesis. Front Oncol 2021; 11:801801. [PMID: 34917515 PMCID: PMC8668865 DOI: 10.3389/fonc.2021.801801] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
The complex and adaptive nature of malignant neoplasm constitute a major challenge for the development of effective anti-oncogenic therapies. Emerging evidence has uncovered the pivotal functions exerted by the small leucine-rich proteoglycans, decorin and biglycan, in affecting tumor growth and progression. In their soluble forms, decorin and biglycan act as powerful signaling molecules. By receptor-mediated signal transduction, both proteoglycans modulate key processes vital for tumor initiation and progression, such as autophagy, inflammation, cell-cycle, apoptosis, and angiogenesis. Despite of their structural homology, these two proteoglycans interact with distinct cell surface receptors and thus modulate distinct signaling pathways that ultimately affect cancer development. In this review, we summarize growing evidence for the complex roles of decorin and biglycan signaling in tumor biology and address potential novel therapeutic implications.
Collapse
Affiliation(s)
- Valentina Diehl
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Lisa Sophie Huber
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung, Member of the German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| |
Collapse
|
11
|
Zhao Y, Li WF, Li QJ, He SW, He QM, Long LF, Liu N, Ma J. WIPI-1 inhibits metastasis and tumour growth via the WIPI-1-TRIM21 axis and MYC regulation in nasopharyngeal carcinoma. Oral Oncol 2021; 122:105576. [PMID: 34689010 DOI: 10.1016/j.oraloncology.2021.105576] [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: 05/08/2021] [Revised: 08/30/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022]
Abstract
The metastatic rate of nasopharyngeal carcinoma (NPC) is the highest among head and neck tumours. Additionally, distant metastasis is the main cause of therapy failure and mortality in NPC. Thus, novel biomarkers are needed for designing new therapeutic strategies to improve the prognosis of this disease. In this study, qRT-PCR and western blotting revealed that the expression of the WD repeat domain phosphoinositide interacting 1 (WIPI-1) was markedly decreased in NPC cells and tissues. Furthermore, low WIPI-1 expression closely correlated with poor prognosis in NPC patients. In vitro functional experiments revealed that overexpression or knockdown of WIPI-1 repressed or facilitated the migration, colony formation, and proliferation of NPC cells. Consistent with the in vitro studies, WIPI-1 significantly inhibited tumour growth, invasion and metastasis in popliteal lymph node metastasis, lung metastasis, and xenograft mouse models in vivo. Mechanistically, WIPI-1 directly interacted with tripartite motif containing 21 (TRIM21) and enhanced starvation-induced autophagy by interacting with TRIM21 in NPC cells. Moreover, MYC gene expression was markedly increased in the WIPI-1 knockdown group, as demonstrated by RNA-seq analysis and qRT-PCR validation. Altogether, WIPI-1 acts as a tumour suppressor gene in NPC that inhibits tumour growth and metastasis. Targeting WIPI-1 may be a novel treatment approach for NPC.
Collapse
Affiliation(s)
- Yin Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China
| | - Wen-Fei Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China
| | - Qing-Jie Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China
| | - Shi-Wei He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China
| | - Qing-Mei He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China
| | - Liu-Fen Long
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China
| | - Na Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China
| | - Jun Ma
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, PR China.
| |
Collapse
|
12
|
Xu T, Xu X, Chu Y, Jiang D, Xu G. Long‑chain non‑coding RNA GAS5 promotes cell autophagy by modulating the miR‑181c‑5p/ ATG5 and miR‑1192/ ATG12 axes. Int J Mol Med 2021; 48:209. [PMID: 34608496 PMCID: PMC8510682 DOI: 10.3892/ijmm.2021.5042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
The main aim of the present study was to explore the role of long-chain non-coding RNA (lncRNA) growth arrest-specific transcript 5 (GAS5) in macrophage autophagy. Firstly, the expression of lncRNA GAS5 during cell starvation or following treatment with 3-methyladenine was determined using reverse transcription-quantitative PCR (RT-qPCR). Additionally, fluorescent in situ hybridization (FISH) assay was utilized to determine the localization of the expression of lncRNA GAS5 in RAW264.7 cells. In vitro cell models were established through the transfection of LV5-lncRNA GAS5 (LV5-GAS5) or LV3-shRNA-lnc GAS5 (sh-GAS5), in order to overexpress or knockdown lncRNA GAS5 expression in RAW264.7 cells. The potential target microRNAs (miRNAs/miRs) of lncRNA GAS5 were analyzed using bioinformatics. The formation of autophagic bodies was detected with the use of laser confocal and transmission electron microscopy. Dual-luciferase reporter assay was performed to determine the target specificities of miR-181c-5p or miR-1192 to lncRNA GAS5 and autophagy-related gene (ATG) or ATG12. The mRNA levels of miR181c-5p, miR-1192, as well as ATG5 and ATG12 were detected using RT-qPCR. The protein levels of microtubule-associated proteins 1A/1B light chain 3B (LC3), p62, ATG5 and ATG12 were measured using western blot analysis. It was revealed that lncRNA GAS5 expression in RAW264.7 macrophages increased significantly during starvation-induced autophagy, and that lncRNA GAS5 overexpression was able to markedly promote the formation of autophagic bodies. Bioinformatics analysis demonstrated that miR-181c-5p and miR-1192 were potential targets of lncRNA GAS5, which was further confirmed by RT-qPCR, western blot analysis and the dual-luciferase reporter assay. Finally, it was confirmed that lncRNA GAS5 promoted autophagy by sponging miR-181c-5p and miR-1192, and upregulating the expression levels of the key autophagic regulators, ATG5 and ATG12. On the whole, the present study demonstrates that total, lncRNA GAS5 promotes macrophage autophagy by targeting the miR-181c-5p/ATG5 and miR-1192/ATG12 axes.
Collapse
Affiliation(s)
- Tao Xu
- Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Xiangrong Xu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yuankui Chu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Dan Jiang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Guangxian Xu
- Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
13
|
Lv D, Chen L, Du L, Zhou L, Tang H. Emerging Regulatory Mechanisms Involved in Liver Cancer Stem Cell Properties in Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:691410. [PMID: 34368140 PMCID: PMC8339910 DOI: 10.3389/fcell.2021.691410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/01/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of primary liver cancer and one of the leading causes of cancer-related deaths worldwide. A growing body of evidence supports the hypothesis that HCC is driven by a population of cells called liver cancer stem cells (LCSCs). LCSCs have been proposed to contribute to malignant HCC progression, including promoting tumor occurrence and growth, mediating tumor metastasis, and treatment resistance, but the regulatory mechanism of LCSCs in HCC remains unclear. Understanding the signaling pathways responsible for LCSC maintenance and survival may provide opportunities to improve patient outcomes. Here, we review the current literature about the origin of LCSCs and the niche composition, describe the current evidence of signaling pathways that mediate LCSC stemness, then highlight several mechanisms that modulate LCSC properties in HCC progression, and finally, summarize the new developments in therapeutic strategies targeting LCSCs markers and regulatory pathways.
Collapse
Affiliation(s)
- Duoduo Lv
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Liyu Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyun Zhou
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Center of Infectious Diseases, Division of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Kocak M, Ezazi Erdi S, Jorba G, Maestro I, Farrés J, Kirkin V, Martinez A, Pless O. Targeting autophagy in disease: established and new strategies. Autophagy 2021; 18:473-495. [PMID: 34241570 PMCID: PMC9037468 DOI: 10.1080/15548627.2021.1936359] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved pathway responsible for clearing cytosolic aggregated proteins, damaged organelles or invading microorganisms. Dysfunctional autophagy leads to pathological accumulation of the cargo, which has been linked to a range of human diseases, including neurodegenerative diseases, infectious and autoimmune diseases and various forms of cancer. Cumulative work in animal models, application of genetic tools and pharmacologically active compounds, has suggested the potential therapeutic value of autophagy modulation in disease, as diverse as Huntington, Salmonella infection, or pancreatic cancer. Autophagy activation versus inhibition strategies are being explored, while the role of autophagy in pathophysiology is being studied in parallel. However, the progress of preclinical and clinical development of autophagy modulators has been greatly hampered by the paucity of selective pharmacological agents and biomarkers to dissect their precise impact on various forms of autophagy and cellular responses. Here, we summarize established and new strategies in autophagy-related drug discovery and indicate a path toward establishing a more efficient discovery of autophagy-selective pharmacological agents. With this knowledge at hand, modern concepts for therapeutic exploitation of autophagy might become more plausible. Abbreviations: ALS: amyotrophic lateral sclerosis; AMPK: AMP-activated protein kinase; ATG: autophagy-related gene; AUTAC: autophagy-targeting chimera; CNS: central nervous system; CQ: chloroquine; GABARAP: gamma-aminobutyric acid type A receptor-associated protein; HCQ: hydroxychloroquine; LYTAC: lysosome targeting chimera; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NDD: neurodegenerative disease; PDAC: pancreatic ductal adenocarcinoma; PE: phosphatidylethanolamine; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; PROTAC: proteolysis-targeting chimera; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; SQSTM1/p62: sequestosome 1; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Muhammed Kocak
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | | | | | - Inés Maestro
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain
| | | | - Vladimir Kirkin
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | - Ana Martinez
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Neurodegenerativas (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - Ole Pless
- Fraunhofer ITMP ScreeningPort, Hamburg, Germany
| |
Collapse
|
15
|
Zhang J, Mao W, Liu Y, Ding J, Wang J, Yu Z, Huang R, Yang S, Sun Y, Dong P. 3-MA Enhanced Chemosensitivity in Cisplatin Resistant Hypopharyngeal Squamous Carcinoma Cells via Inhibiting Beclin -1 Mediated Autophagy. Curr Pharm Des 2021; 27:996-1005. [PMID: 33349212 DOI: 10.2174/1381612826666201221150431] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hypopharyngeal carcinoma is characterized by a high degree of malignancy. The most common pathological type is squamous cell carcinoma (HSCC). Cisplatin (cis-diamminedichloroplatinum, CDDP) is one of the most widely used chemotherapeutic drugs nowadays and cisplatin resistance is a major problem in current treatment strategies. Clinical researchers have reported that high autophagy levels often caused insensitivity to chemotherapy, a common phenomenon that greatly reduces the therapeutic effect in cisplatin- resistant tumor cell lines. 3-methyladenine (3-MA), an inhibitor of PI3K, plays a vital role in forming and developing autophagosomes. Therefore, we speculate that the use of 3-MA may reduce cisplatin resistance in hypopharyngeal squamous cell carcinoma (HSCC). METHODS Part I: Cisplatin-resistant FaDu cell line (Human hypopharyngeal squamous cell carcinoma cells) was established and cultured. Cell counting kit-8 was used to detect drug resistance. An inverted microscope was used to observe the morphological changes at different concentrations, then the survival rate was calculated. After MDC staining, the autophagic vacuoles were observed by fluorescence microscopy. The expression of Beclin1 from each group was confirmed by RT-PCR and Western blot method. Part II: 3-MA was applied for cisplatin-resistant cells intervention, Beclin1 was knocked down by plasmid transfection. Cell cycle was detected using flow cytometry assay, apoptosis with necrosis was detected by staining with propidium iodide (PI). CCK-8 was used to observe the cell survival rate in each group. The expression of autophagy-related protein Beclin1, LC3I, LC3II, Atg-5 and P62 in each group was verified by Western blot analysis. RESULTS Cisplatin-resistant FaDu cell line can be stably constructed by cisplatin intervention. Compared with normal group, autophagy and its related protein Beclin1 expression were enhanced in cisplatin resistant FaDu cells. Autophagy inhibition group showed significant cell cycle changes, mainly manifested by G1 arrest, increased apoptosis rate and significantly decreased survival rate at 24h level. The number of autophagy vacuoles were significantly reduced in the 3-MA group. Furthermore, Western blot showed that expression of Beclin1, lc3-I, lc3-II, atg-5 protein decreased significantly after 3-MA intervention, while the expression of p62 upregulated, which also confirmed autophagy flow was blocked. CONCLUSION Our work confirmed that enhanced autophagy is an important cause of cisplatin resistance in FaDu cells. The use of 3-MA can significantly reduce autophagy level and arresting its cell cycle, promote apoptosis and reverse the cisplatin resistance condition, this effect is partly mediated by inhibition of Beclin-1 expression. Our data provide a theoretical basis for the application of 3-MA in overcoming cisplatin resistance in hypopharyngeal cancer.
Collapse
Affiliation(s)
- Jia Zhang
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Wei Mao
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Yuying Liu
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Jian Ding
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Jie Wang
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Ziwei Yu
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Ruofei Huang
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Sen Yang
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Ying Sun
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| | - Pin Dong
- Division of ENT & HN Surgery, Shanghai General Hospital of Nanjing Medical University, Key Laboratory of Head and Neck, Shanghai, 200080, China
| |
Collapse
|
16
|
Rosenfeldt MT, O'Prey J, Lindsay CR, Nixon C, Roth S, Sansom OJ, Ryan KM. Loss of autophagy affects melanoma development in a manner dependent on PTEN status. Cell Death Differ 2021; 28:1437-1439. [PMID: 33664481 PMCID: PMC8027884 DOI: 10.1038/s41418-021-00746-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/15/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Mathias T Rosenfeldt
- Cancer Research UK Beatson Institute, Glasgow, UK.
- Comprehensive Cancer Center Mainfranken, Wuerzburg, Germany.
- Department of Pathology, University of Wuerzburg, Wuerzburg, Germany.
| | - Jim O'Prey
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Colin Nixon
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Sabine Roth
- Department of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
17
|
Zhang Z, Zhang Y, Mo W. The Autophagy Related Gene CHAF1B Is a Relevant Prognostic and Diagnostic Biomarker in Hepatocellular Carcinoma. Front Oncol 2021; 10:626175. [PMID: 33575221 PMCID: PMC7870991 DOI: 10.3389/fonc.2020.626175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/14/2020] [Indexed: 12/01/2022] Open
Abstract
The role of autophagy in tumors is complex; based on known interactions between autophagy and hepatocellular carcinoma (HCC) pathogenesis, we hypothesized that autophagy-related genes (ARGs) may play an important role in HCC. The ARGs were obtained from the Human Autophagy Database and the Gene Set Enrichment Analysis. Based on the area under the curve (AUC) value >0.9 with p <0.0001 and Student's T-test analysis with p <0.0001, differently expressed autophagy-related genes (DEARGs) with high diagnostic efficiency were found. Besides that, we searched in the PubMed database to find novel DEARGs associated with HCC. Then the DEARGs were validated in the GSE25097, GSE54236, GSE76427, GSE64041, Oncomine, and Human Protein Atlas datasets. Finally, survival analysis of CHAF1B in HCC and correlations of clinico-pathological characteristics and CHAF1B were performed based on the TCGA database. The mRNA and protein expression of 531 ARGs were analyzed and validated in eight independent cohorts. First, 18 DEARGs with high diagnostic efficiency were selected from the TCGA database, and nine of them were identified that had not previously been associated with HCC. These nine DEARGs were validated in the GSE25097, GSE54236, GSE76427, GSE64041, Oncomine, and Human Protein Atlas datasets. Additionally, we found that CHAF1B was associated with overall survival and relapse free survival at one, three, and five years. Furthermore, the univariate and multivariate Cox analyses revealed that the high expression of CHAF1B was an independent risk factor in HCC patients. This research demonstrated that CHAF1B was a novel diagnostic and prognostic signature biomarker that could be potentially useful for predicting the development of HCC and may provide new insights for HCC tumorigenesis and treatments.
Collapse
Affiliation(s)
- Zunni Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yalong Zhang
- Department of Ultrasonic Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wuning Mo
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
18
|
Xue L, Liu P. Daurisoline inhibits hepatocellular carcinoma progression by restraining autophagy and promoting cispaltin-induced cell death. Biochem Biophys Res Commun 2021; 534:1083-1090. [PMID: 33213840 DOI: 10.1016/j.bbrc.2020.09.068] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy with high cancer-associated mortality. Suppressing autophagy has been reported to promote the efficiency of chemotherapy in HCC. Daurisoline (DAS) is a constituent of Rhizoma Menispermi, and functions as a potential autophagy inhibitor to perform different cellular events. In the present study, we found that DAS treatment up-regulated autophagosomes in HCC cells, accompanied with the increases of LC3-II and p62, demonstrating the disturbance of autophagic flux. Then, by the colocalization analysis, we identified that DAS did not repress the fusion of autophagosomes and lysosomes in HCC cells. However, Lysotracker and acridine orange (OA) staining showed that DAS could suppress lysosomal acidification, as evidenced by the decreased red fluorescence. Consistently, significant decreases in mature form of cathepsin B and cathepsin D were detected in DAS-treated HCC cells. Furthermore, DAS treatment markedly promoted the anti-cancer effects of cisplatin (cDDP) on HCC cells, as revealed by the dramatically reduced cell viability and proliferation, whereas the enhanced apoptosis. Moreover, the nude mice xenograft models with HCC confirmed that compared with cDDP alone group, DAS combined with cDDP significantly reduced tumor progression in vivo. Taken together, these findings elucidated that DAS could restrain autophagic flux, potentiating the chemosensitivity of HCC cells to cDDP treatment.
Collapse
Affiliation(s)
- Legang Xue
- Department of Pharmacy, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Pei Liu
- Department of Pharmacy, The Fourth People's Hospital of Huai'an, Huai'an, 223002, China.
| |
Collapse
|
19
|
The Dual Role of Autophagy in Cancer Development and a Therapeutic Strategy for Cancer by Targeting Autophagy. Int J Mol Sci 2020; 22:ijms22010179. [PMID: 33375363 PMCID: PMC7795059 DOI: 10.3390/ijms22010179] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a delicate intracellular degradation process that occurs due to diverse stressful conditions, including the accumulation of damaged proteins and organelles as well as nutrient deprivation. The mechanism of autophagy is initiated by the creation of autophagosomes, which capture and encapsulate abnormal components. Afterward, autophagosomes assemble with lysosomes to recycle or remove degradative cargo. The regulation of autophagy has bipolar roles in cancer suppression and promotion in diverse cancers. Furthermore, autophagy modulates the features of tumorigenesis, cancer metastasis, cancer stem cells, and drug resistance against anticancer agents. Some autophagy regulators are used to modulate autophagy for anticancer therapy but the dual roles of autophagy limit their application in anticancer therapy, and present as the main reason for therapy failure. In this review, we summarize the mechanisms of autophagy, tumorigenesis, metastasis, cancer stem cells, and resistance against anticancer agents. Finally, we discuss whether targeting autophagy is a promising and effective therapeutic strategy in anticancer therapy.
Collapse
|
20
|
Akhtar MF, Saleem A, Rasul A, Faran Ashraf Baig MM, Bin-Jumah M, Abdel Daim MM. Anticancer natural medicines: An overview of cell signaling and other targets of anticancer phytochemicals. Eur J Pharmacol 2020; 888:173488. [PMID: 32805253 DOI: 10.1016/j.ejphar.2020.173488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/23/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Therapies of cancer are as diverse as multifaceted the cancer is. Anticancer drugs include, but not limited to synthetic, semisynthetic and natural drugs and monoclonal antibodies. A recent decline in new drug development has led to the rebirth of herbal therapeutics in the form of dietary supplements and botanical preparations. Medicinal plants comprise of complex phytochemicals due to vast biosynthetic capacity. A wide array of phytochemicals has been pharmacologically evaluated for their chemo-preventive and chemotherapeutic potential for several decades. These phytochemicals target cancer at diverse sites such as apoptotic pathways, genetic and epigenetic mutations, damage to deoxyribonucleic acid, production of reactive oxygen species, autophagy, invasion and metastasis of cancer cells, and modulation of cell signaling through Janus-activated kinase/Signal transducer and activator of transcription, Notch, mitogen-activated protein kinase/Extracellular signal-regulated kinase, phosphatidylinositol 3-kinase/Protein kinase B/mammalian target of rapamycin, Nuclear factor kappa B, Wingless-related integration site and Transforming growth factor β pathways. This review focuses on the therapeutic targets of anticancer and chemo-preventive phytochemicals and their mode of action.
Collapse
Affiliation(s)
- Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Pakistan.
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan
| | | | - May Bin-Jumah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M Abdel Daim
- Department of Zoology, College of Science, King Saud University, 2455, Riyadh, 11451, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
21
|
Long M, McWilliams TG. Monitoring autophagy in cancer: From bench to bedside. Semin Cancer Biol 2020; 66:12-21. [DOI: 10.1016/j.semcancer.2019.05.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/06/2019] [Accepted: 05/27/2019] [Indexed: 12/29/2022]
|
22
|
Chiu CT, Lin CY, Yen CY, Tsai MT, Chang HC, Liu YC, Lin MH. Mechanistic and compositional studies of the autophagy-inducing areca nut ingredient. J Dent Sci 2020; 15:526-535. [PMID: 33505626 PMCID: PMC7816039 DOI: 10.1016/j.jds.2020.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/03/2020] [Indexed: 12/13/2022] Open
Abstract
Background/purpose We previously found that the partially purified 30–100 kDa fraction of areca-nut-extract (ANE 30–100K) induces autophagy in different types of cells including oral carcinoma OECM-1 cells. This study was to analyze the composition and possible mechanisms of ANE 30-100K-induced autophagy (AIA). Materials and methods Phenol-sulfuric acid method and high performance anion exchange chromatography were utilized to analyze the composition of ANE 30–100K. OECM-1 and esophageal CE81T/VGH cells were taken as the experimental models. Microscope and transmission electron microscope were used to observe morphological changes. Cell viability and specific proteins were respectively measured by XTT and Western bot assay. shRNA and chemical inhibitors were applied to assess the involvement of Atg5, caveolin, and proteasome in AIA. Results ANE 30–100K contains ∼67% carbohydrate, which is composed of fucose (5.938%), arabinose (24.631%), glucosamine (8.066%), galactose (26.820%), glucose (21.388%), and mannose (13.157%). After ANE 30–100K stimulation, CE81T/VGH cells showed intracellular vacuoles, acidic vesicles, double-membrane vacuoles, and elevated LC3-II level. ANE 30-100K-induced cytotoxicity and LC3-II accumulation were significantly inhibited by Atg5 knockdown. Furthermore, the endocytosis inhibitor (methyl-β-cyclodextrin) and two caveolin shRNAs, as well as two proteasome inhibitors (lactacystin and epoxomicin), were shown to significantly attenuate ANE 30-100K-induced cytotoxicity and LC3-II accumulation in both OECM-1 and CE81T/VGH cells. Conclusion The major components of ANE 30–100K are carbohydrates. CE81T/VGH also exhibited autophagic responses to ANE 30–100K. Caveolin-mediated endocytosis and proteasome are involved in AIA. This study may have provided new knowledges of the action mechanisms and compositions of ANE 30–100K.
Collapse
Affiliation(s)
- Chang-Ta Chiu
- Department of Dentistry, Tainan Municipal An-Nan Hospital China Medical University, Tainan, Taiwan, Republic of China
| | - Che-Yi Lin
- Oral and Maxillofacial Surgery Section, Chi Mei Medical Center, Tainan, Taiwan, Republic of China
| | - Ching-Yu Yen
- Oral and Maxillofacial Surgery Section, Chi Mei Medical Center, Tainan, Taiwan, Republic of China.,Department of Dentistry, Taipei Medical University, Taipei, Taiwan, Republic of China.,Department of Dentistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Meng-Ting Tsai
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan, Republic of China
| | - Huei-Cih Chang
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan, Republic of China
| | - Young-Chau Liu
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, Republic of China.,Division of Natural Science, College of Liberal Education, Shu-Te University, Yanchao District, Kaohsiung City, Taiwan, Republic of China
| | - Mei-Huei Lin
- Department of Biotechnology, Chia Nan University of Pharmacy, Tainan, Taiwan, Republic of China.,Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, Republic of China
| |
Collapse
|
23
|
Crowley E, Leung E, Reynisson J, Richardson A. Rapid changes in the ATG5-ATG16L1 complex following nutrient deprivation measured using NanoLuc Binary Technology (NanoBIT). FEBS J 2020; 287:4917-4932. [PMID: 32129924 DOI: 10.1111/febs.15275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 01/16/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022]
Abstract
Autophagy plays a role in several human diseases, but each of the current methods to measure autophagy has significant drawbacks. ATG5 and ATG16L1 are regulators necessary for autophagy; therefore, drugs that inhibit the interaction of these proteins may be therapeutically useful. To evaluate the interaction of ATG5 and ATG16L1 in cells, their cDNAs were fused to the coding sequences of SmBIT and LgBIT, two components of NanoLuc luciferase. This generated a luminescent signal when SmBIT and LgBIT interacted to form a functional luciferase as a result of their colocalization that was brought about by the binding of ATG5 and ATG16L1. The assay measures the interaction in real time and can be used in microplate format to allow for multiple experimental conditions to be assessed. The interaction of ATG5 and ATG16L1 is not significantly altered by inhibition of lysosomal function, or inhibitors of Ulk1, Vps34 or mTORC1. However, there was a constitutive interaction of ATG5 and ATG16L1 and luminescence was stimulated within 3 min, by up to 500%, when the cells were deprived of nutrients. When the nutrients are returned, the complex returns to its basal status equally rapidly. Sphingosine-1-phosphate and CYM-5541 partially repressed the effects of nutrient starvation. Furthermore, we identified a small-molecule inhibitor that interferes with the interaction of ATG5 and ATG16L1 in cells. This assay provides a novel tool for researchers to measure autophagy and can be potentially applied to many cell types. DATABASE: Replicate data are available in Figshare database https://doi.org/10.6084/m9.figshare.11798946.
Collapse
Affiliation(s)
- Emily Crowley
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Stoke-on-Trent, UK
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Jóhannes Reynisson
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Stoke-on-Trent, UK
| | - Alan Richardson
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Stoke-on-Trent, UK
| |
Collapse
|
24
|
Rojas-Sanchez G, Cotzomi-Ortega I, Pazos-Salazar NG, Reyes-Leyva J, Maycotte P. Autophagy and Its Relationship to Epithelial to Mesenchymal Transition: When Autophagy Inhibition for Cancer Therapy Turns Counterproductive. BIOLOGY 2019; 8:biology8040071. [PMID: 31554173 PMCID: PMC6956138 DOI: 10.3390/biology8040071] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/14/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023]
Abstract
The manipulation of autophagy for cancer therapy has gained recent interest in clinical settings. Although inhibition of autophagy is currently being used in clinical trials for the treatment of several malignancies, autophagy has been shown to have diverse implications for normal cell homeostasis, cancer cell survival, and signaling to cells in the tumor microenvironment. Among these implications and of relevance for cancer therapy, the autophagic process is known to be involved in the regulation of protein secretion, in tumor cell immunogenicity, and in the regulation of epithelial-to-mesenchymal transition (EMT), a critical step in the process of cancer cell invasion. In this work, we have reviewed recent evidence linking autophagy to the regulation of EMT in cancer and normal epithelial cells, and have discussed important implications for the manipulation of autophagy during cancer therapy.
Collapse
Affiliation(s)
- Guadalupe Rojas-Sanchez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Israel Cotzomi-Ortega
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Nidia G Pazos-Salazar
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
| | - Julio Reyes-Leyva
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Paola Maycotte
- Consejo Nacional de Ciencia y Tecnología (CONACYT)-CIBIOR, IMSS, Puebla 74360, Mexico.
| |
Collapse
|
25
|
Zhang Y, Xie Y, Liu W, Deng W, Peng D, Wang C, Xu H, Ruan C, Deng Y, Guo Y, Lu C, Yi C, Ren J, Xue Y. DeepPhagy: a deep learning framework for quantitatively measuring autophagy activity in Saccharomyces cerevisiae. Autophagy 2019; 16:626-640. [PMID: 31204567 DOI: 10.1080/15548627.2019.1632622] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Seeing is believing. The direct observation of GFP-Atg8 vacuolar delivery under confocal microscopy is one of the most useful end-point measurements for monitoring yeast macroautophagy/autophagy. However, manually labelling individual cells from large-scale sets of images is time-consuming and labor-intensive, which has greatly hampered its extensive use in functional screens. Herein, we conducted a time-course analysis of nitrogen starvation-induced autophagy in wild-type and knockout mutants of 35 AuTophaGy-related (ATG) genes in Saccharomyces cerevisiae and obtained 1,944 confocal images containing > 200,000 cells. We manually labelled 8,078 autophagic and 18,493 non-autophagic cells as a benchmark dataset and developed a new deep learning tool for autophagy (DeepPhagy), which exhibited superior accuracy in recognizing autophagic cells compared to other existing methods, with an area under the curve (AUC) value of 0.9710 from 10-fold cross-validations. We further used DeepPhagy to automatically analyze all the images and quantitatively classified the autophagic phenotypes of the 35 atg knockout mutants into 3 classes. The high consistency in our computational and biochemical results indicated the reliability of DeepPhagy for measuring autophagic activity. Moreover, we used DeepPhagy to analyze 3 additional types of autophagic phenotypes, including the targeting of Atg1-GFP to the vacuole, the vacuolar delivery of GFP-Atg19, and the disintegration of autophagic bodies indicated by GFP-Atg8, all with satisfying accuracies. Taken together, our study not only enables the GFP-Atg8 fluorescence assay to become a quantitative measurement for analyzing autophagic phenotypes in S. cerevisiae but also demonstrates that deep learning-based methods could potentially be applied to different types of autophagy.Abbreviations: Ac: accuracy; ALP: alkaline phosphatase; ALR: autophagic lysosomal reformation; ATG: AuTophaGy-related; AUC: area under the curve; CNN: convolutional neural network; Cvt: cytoplasm-to-vacuole targeting; DeepPhagy: deep learning for autophagy; fc_2: second fully connected; GFP: green fluorescent protein; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3 beta; HAT: histone acetyltransferase; HemI: Heat map Illustrator; JRE: Java Runtime Environment; KO: knockout; LRN: local response normalization; MCC: Mathew Correlation Coefficient; OS: operating system; PAS: phagophore assembly site; PC: principal component; PCA: principal component analysis; PPI: protein-protein interaction; Pr: precision; QPSO: Quantum-behaved Particle Swarm Optimization; ReLU: rectified linear unit; RF: random forest; ROC: receiver operating characteristic; ROI: region of interest; SD: systematic derivation; SGD: stochastic gradient descent; Sn: sensitivity; Sp: specificity; SRG: seeded region growing; t-SNE: t-distributed stochastic neighbor embedding; 2D: 2-dimensional; WT: wild-type.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yubin Xie
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenzhong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wankun Deng
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Di Peng
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chenwei Wang
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Haodong Xu
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Ruan
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yongjie Deng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yaping Guo
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chenjun Lu
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong Yi
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Ren
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Xue
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Sharma G, Guardia CM, Roy A, Vassilev A, Saric A, Griner LN, Marugan J, Ferrer M, Bonifacino JS, DePamphilis ML. A family of PIKFYVE inhibitors with therapeutic potential against autophagy-dependent cancer cells disrupt multiple events in lysosome homeostasis. Autophagy 2019; 15:1694-1718. [PMID: 30806145 DOI: 10.1080/15548627.2019.1586257] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
High-throughput screening identified 5 chemical analogs (termed the WX8-family) that disrupted 3 events in lysosome homeostasis: (1) lysosome fission via tubulation without preventing homotypic lysosome fusion; (2) trafficking of molecules into lysosomes without altering lysosomal acidity, and (3) heterotypic fusion between lysosomes and autophagosomes. Remarkably, these compounds did not prevent homotypic fusion between lysosomes, despite the fact that homotypic fusion required some of the same machinery essential for heterotypic fusion. These effects varied 400-fold among WX8-family members, were time and concentration dependent, reversible, and resulted primarily from their ability to bind specifically to the PIKFYVE phosphoinositide kinase. The ability of the WX8-family to prevent lysosomes from participating in macroautophagy/autophagy suggested they have therapeutic potential in treating autophagy-dependent diseases. In fact, the most potent family member (WX8) was 100-times more lethal to 'autophagy-addicted' melanoma A375 cells than the lysosomal inhibitors hydroxychloroquine and chloroquine. In contrast, cells that were insensitive to hydroxychloroquine and chloroquine were also insensitive to WX8. Therefore, the WX8-family of PIKFYVE inhibitors provides a basis for developing drugs that could selectively kill autophagy-dependent cancer cells, as well as increasing the effectiveness of established anti-cancer therapies through combinatorial treatments. Abbreviations: ACTB: actin beta; Baf: bafilomycin A1; BECN1: beclin 1; BODIPY: boron-dipyrromethene; BORC: BLOC-1 related complex; BRAF: B-Raf proto-oncogene, serine/threonine kinase; BSA: bovine serum albumin; CTSD: cathepsin D; CQ: chloroquine; DNA: deoxyribonucleic acid; EC50: half maximal effective concentration; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HCQ: hydroxychloroquine; HOPS complex: homotypic fusion and protein sorting complex; Kd: equilibrium binding constant; IC50: half maximal inhibitory concentration; KO: knockout; LAMP1: lysosomal associated membrane protein 1; MAP1LC3A: microtubule associated protein 1 light chain 3 alpha; MES: 2-(N-morpholino)ethanesulphonic acid; MTOR: mechanistic target of rapamycin kinase; μM: micromolar; NDF: 3-methylbenzaldehyde (2,6-dimorpholin-4-ylpyrimidin-4-yl)hydrazine;NEM: N-ethylmaleimide; NSF: N-ethylmaleimide sensitive factor; PBS: phosphate-buffered saline; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PIP4K2C: phosphatidylinositol-5-phosphate 4-kinase type 2 gamma; PtdIns3P: phosphatidylinositol 3-phosphate; PtdIns(3,5)P2: phosphatidylinositol 3,5-biphosphate; RFP: red fluorescent protein; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; SQSTM1: sequestosome 1; TWEEN 20: polysorbate 20; V-ATPase: vacuolar-type H+-translocating ATPase; VPS39: VPS39 subunit of HOPS complex; VPS41: VPS41 subunit of HOPS complex; WWL: benzaldehyde [2,6-di(4-morpholinyl)-4-pyrimidinyl]hydrazone; WX8: 1H-indole-3-carbaldehyde [4-anilino-6-(4-morpholinyl)-1,3,5-triazin-2-yl]hydrazine; XBA: N-(3-chloro-4-fluorophenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride; XB6: N-(4-ethylphenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride.
Collapse
Affiliation(s)
- Gaurav Sharma
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Carlos M Guardia
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Ajit Roy
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Alex Vassilev
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Amra Saric
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Lori N Griner
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Juan Marugan
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Marc Ferrer
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Melvin L DePamphilis
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
27
|
Nazio F, Bordi M, Cianfanelli V, Locatelli F, Cecconi F. Autophagy and cancer stem cells: molecular mechanisms and therapeutic applications. Cell Death Differ 2019; 26:690-702. [PMID: 30728463 PMCID: PMC6460398 DOI: 10.1038/s41418-019-0292-y] [Citation(s) in RCA: 287] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
Autophagy and mitophagy act in cancer as bimodal processes, whose differential functions strictly depend on cancer ontogenesis, progression, and type. For instance, they can act to promote cancer progression by helping cancer cells survive stress or, instead, when mutated or abnormal, to induce carcinogenesis by influencing cell signaling or promoting intracellular toxicity. For this reason, the study of autophagy in cancer is the main focus of many researchers and several clinical trials are already ongoing to manipulate autophagy and by this way determine the outcome of disease therapy. Since the establishment of the cancer stem cell (CSC) theory and the discovery of CSCs in individual cancer types, autophagy and mitophagy have been proposed as key mechanisms in their homeostasis, dismissal or spread, even though we still miss a comprehensive view of how and by which regulatory molecules these two processes drive cell fate. In this review, we will dive into the deep water of autophagy, mitophagy, and CSCs and offer novel viewpoints on possible therapeutic strategies, based on the modulation of these degradative systems.
Collapse
Affiliation(s)
- Francesca Nazio
- Department of Oncohaematology and Cellular and Gene Therapy, IRCSS Bambino Gesù Children's Hospital, 00165, Rome, Italy
| | - Matteo Bordi
- Department of Oncohaematology and Cellular and Gene Therapy, IRCSS Bambino Gesù Children's Hospital, 00165, Rome, Italy
- Department of Biology, University of Tor Vergata, 00133, Rome, Italy
| | - Valentina Cianfanelli
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Franco Locatelli
- Department of Oncohaematology and Cellular and Gene Therapy, IRCSS Bambino Gesù Children's Hospital, 00165, Rome, Italy
- Department of Gynecology/Obstetrics and Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Francesco Cecconi
- Department of Oncohaematology and Cellular and Gene Therapy, IRCSS Bambino Gesù Children's Hospital, 00165, Rome, Italy.
- Department of Biology, University of Tor Vergata, 00133, Rome, Italy.
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, 2100, Copenhagen, Denmark.
| |
Collapse
|
28
|
WIPI1, BAG1, and PEX3 Autophagy-Related Genes Are Relevant Melanoma Markers. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1471682. [PMID: 30622661 PMCID: PMC6304818 DOI: 10.1155/2018/1471682] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/09/2018] [Indexed: 01/07/2023]
Abstract
ROS and oxidative stress may promote autophagy; on the other hand, autophagy may help reduce oxidative damages. According to the known interplay of ROS, autophagy, and melanoma onset, we hypothesized that autophagy-related genes (ARGs) may represent useful melanoma biomarkers. We therefore analyzed the gene and protein expression of 222 ARGs in human melanoma samples, from 5 independent expression databases (overall 572 patients). Gene expression was first evaluated in the GEO database. Forty-two genes showed extremely high ability to discriminate melanoma from nevi (63 samples) according to ROC (AUC ≥ 0.85) and Mann-Whitney (p < 0.0001) analyses. The 9 genes never related to melanoma before were then in silico validated in the IST online database. BAG1, CHMP2B, PEX3, and WIPI1 confirmed a strong differential gene expression, in 355 samples. A second-round validation performed on the Human Protein Atlas database showed strong differential protein expression for BAG1, PEX3, and WIPI1 in melanoma vs control samples, according to the image analysis of 80 human histological sections. WIPI1 gene expression also showed a significant prognostic value (p < 0.0001) according to 102 melanoma patients' survival data. We finally addressed in Oncomine database whether WIPI1 overexpression is melanoma-specific. Within more than 20 cancer types, the most relevant WIPI1 expression change (p = 0.00002; fold change = 3.1) was observed in melanoma. Molecular/functional relationships of the investigated molecules with melanoma and their molecular/functional network were analyzed via Chilibot software, STRING analysis, and gene ontology enrichment analysis. We conclude that WIPI1 (AUC = 0.99), BAG1 (AUC = 1), and PEX3 (AUC = 0.93) are relevant novel melanoma markers at both gene and protein levels.
Collapse
|
29
|
Limpert AS, Lambert LJ, Bakas NA, Bata N, Brun SN, Shaw RJ, Cosford NDP. Autophagy in Cancer: Regulation by Small Molecules. Trends Pharmacol Sci 2018; 39:1021-1032. [PMID: 30454769 PMCID: PMC6349222 DOI: 10.1016/j.tips.2018.10.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023]
Abstract
During times of stress, autophagy is a cellular process that enables cells to reclaim damaged components by a controlled recycling pathway. This mechanism for cellular catabolism is dysregulated in cancer, with evidence indicating that cancer cells rely on autophagy in the hypoxic and nutrient-poor microenvironment of solid tumors. Mounting evidence suggests that autophagy has a role in the resistance of tumors to standard-of-care (SOC) therapies. Therefore, there is significant interest in the discovery of small molecules that can safely modulate autophagy. In this review, we describe recent advances in the identification of new pharmacological compounds that modulate autophagy, with a focus on their mode of action, value as probe compounds, and validation as potential therapeutics.
Collapse
Affiliation(s)
- Allison S Limpert
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA; These authors contributed equally
| | - Lester J Lambert
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA; These authors contributed equally
| | - Nicole A Bakas
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Nicole Bata
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sonja N Brun
- Department of Molecular and Cell Biology, The Salk Institute for Biological Studies, San Diego, La Jolla, CA, USA
| | - Reuben J Shaw
- Department of Molecular and Cell Biology, The Salk Institute for Biological Studies, San Diego, La Jolla, CA, USA
| | - Nicholas D P Cosford
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
30
|
Kalyanaraman B, Cheng G, Hardy M, Ouari O, Bennett B, Zielonka J. Teaching the basics of reactive oxygen species and their relevance to cancer biology: Mitochondrial reactive oxygen species detection, redox signaling, and targeted therapies. Redox Biol 2017; 15:347-362. [PMID: 29306792 PMCID: PMC5756055 DOI: 10.1016/j.redox.2017.12.012] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/21/2017] [Accepted: 12/23/2017] [Indexed: 01/05/2023] Open
Abstract
Reactive oxygen species (ROS) have been implicated in tumorigenesis (tumor initiation, tumor progression, and metastasis). Of the many cellular sources of ROS generation, the mitochondria and the NADPH oxidase family of enzymes are possibly the most prevalent intracellular sources. In this article, we discuss the methodologies to detect mitochondria-derived superoxide and hydrogen peroxide using conventional probes as well as newly developed assays and probes, and the necessity of characterizing the diagnostic marker products with HPLC and LC-MS in order to rigorously identify the oxidizing species. The redox signaling roles of mitochondrial ROS, mitochondrial thiol peroxidases, and transcription factors in response to mitochondria-targeted drugs are highlighted. ROS generation and ROS detoxification in drug-resistant cancer cells and the relationship to metabolic reprogramming are discussed. Understanding the subtle role of ROS in redox signaling and in tumor proliferation, progression, and metastasis as well as the molecular and cellular mechanisms (e.g., autophagy) could help in the development of combination therapies. The paradoxical aspects of antioxidants in cancer treatment are highlighted in relation to the ROS mechanisms in normal and cancer cells. Finally, the potential uses of newly synthesized exomarker probes for in vivo superoxide and hydrogen peroxide detection and the low-temperature electron paramagnetic resonance technique for monitoring oxidant production in tumor tissues are discussed.
Collapse
Affiliation(s)
- Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States; Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States; Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States.
| | - Gang Cheng
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States; Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Micael Hardy
- Aix Marseille Univ CNRS ICR UMR 7273, Marseille 13013, France
| | - Olivier Ouari
- Aix Marseille Univ CNRS ICR UMR 7273, Marseille 13013, France
| | - Brian Bennett
- Department of Physics, Marquette University, 540 North 15th Street, Milwaukee, WI 53233, United States
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States; Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States; Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| |
Collapse
|
31
|
The Long Noncoding RNA HOTAIR in Breast Cancer: Does Autophagy Play a Role? Int J Mol Sci 2017; 18:ijms18112317. [PMID: 29469819 PMCID: PMC5713286 DOI: 10.3390/ijms18112317] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 01/17/2023] Open
Abstract
HOTAIR (HOX transcript antisense RNA) plays a critical role in chromatin dynamics through the interaction with histone modifiers resulting in transcriptional gene silencing. The promoter of the HOTAIR gene contains multiple estrogen response elements (EREs) and is transcriptionally activated by estradiol in estrogen receptor-positive breast cancer cells. HOTAIR competes with BRCA1, a critical protein in breast cancer and is a critical regulator of genes involved in epithelial-to-mesenchymal transition. It mediates an oncogenic action of c-Myc, essential for breast carcinogenesis. The carcinogenic action of HOTAIR was confirmed in breast cancer stem-like cells, in which it was essential for self-renewal and proliferation. Several miRNAs regulate the expression of HOTAIR and HOTAIR interacts with many miRNAs to support cancer transformation. Many studies point at miR-34a as a major component of HOTAIR–miRNAs–cancer cross-talk. The most important role of HOTAIR can be attributed to cancer progression as its overexpression stimulates invasion and metastasis. HOTAIR can regulate autophagy, important for breast cancer cells survival, through the interaction with miRNAs specific for autophagy genes and directly with these genes. The role of HOTAIR-mediated autophagy in breast cancer progression can be underlined by its interaction with matrix metalloproteinases, essential for cancer invasion, and β-catenin can be important for this interaction. Therefore, there are several mechanisms of the interplay between HOTAIR and autophagy important for breast cancer, but further studies are needed to determine more details of this interplay.
Collapse
|