1
|
Liu H, Su P, Li Y, Hoover A, Hu S, King SA, Zhao J, Guan JL, Chen SY, Zhao Y, Tan M, Wu X. VAMP2 controls murine epidermal differentiation and carcinogenesis by regulation of nucleophagy. Dev Cell 2024; 59:2005-2016.e4. [PMID: 38810653 PMCID: PMC11303110 DOI: 10.1016/j.devcel.2024.05.004] [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: 09/26/2023] [Revised: 02/16/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
Differentiation of murine epidermal stem/progenitor cells involves the permanent withdrawal from the cell cycle, the synthesis of various protein and lipid components for the cornified envelope, and the controlled dissolution of cellular organelles and nuclei. Deregulated epidermal differentiation contributes to the development of various skin diseases, including skin cancers. With a genome-wide shRNA screen, we identified vesicle-associated membrane protein 2 (VAMP2) as a critical factor involved in skin differentiation. Deletion of VAMP2 leads to aberrant skin stratification and enucleation in vivo. With quantitative proteomics, we further identified an autophagy protein, focal adhesion kinase family interacting protein of 200 kDa (FIP200), as a binding partner of VAMP2. Additionally, we showed that both VAMP2 and FIP200 are critical for murine keratinocyte enucleation and epidermal differentiation. Loss of VAMP2 or FIP200 enhances cutaneous carcinogenesis in vivo. Together, our findings identify important molecular mechanisms underlying epidermal differentiation and skin tumorigenesis.
Collapse
Affiliation(s)
- Han Liu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Peihong Su
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Yuanyuan Li
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Alex Hoover
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Sophie Hu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Sarah A King
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Jing Zhao
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Yingming Zhao
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Minjia Tan
- The Chemical Proteomics Center and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Peralta J, DuPriest B, Orozco D, Pacheco JR, Martins L, Soriano RA, Wong A, Wong R, Grillo-Hill B. Drosophila Nhe2 overexpression induces autophagic cell death. Mol Biol Cell 2024; 35:br13. [PMID: 38696256 PMCID: PMC11244158 DOI: 10.1091/mbc.e24-02-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/04/2024] Open
Abstract
Autophagy is a conserved catabolic process where double membrane-bound structures form around macromolecules or organelles targeted for degradation. Autophagosomes fuse with lysosomes to facilitate degradation and macromolecule recycling for homeostasis or growth in a cell autonomous manner. In cancer cells, autophagy is often up-regulated and helps cancer cells survive nutrient deprivation and stressful growth conditions. Here, we propose that the increased intracellular pH (pHi) common to cancer cells is sufficient to induce autophagic cell death. We previously developed tools to increase pHi in the Drosophila eye via overexpression of DNhe2, resulting in aberrant patterning and reduced tissue size. We examined fly eyes at earlier stages of development and found fewer interommatidial cells. We next tested whether this decrease in cell number was due to increased cell death. We found that the DNhe2-induced cell death was caspase independent, which is inconsistent with apoptosis. However, this cell death required autophagy genes, which supports autophagy as the mode of cell death. We also found that expression of molecular markers supports increased autophagy. Together, our findings suggest new roles for ion transport proteins in regulating conserved, critical developmental processes and provide evidence for new paradigms in growth control.
Collapse
Affiliation(s)
- Jobelle Peralta
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Blake DuPriest
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Daniel Orozco
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Juan Reyna Pacheco
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Laura Martins
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Rachel Ann Soriano
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Alan Wong
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Ramy Wong
- Department of Biological Sciences, San José State University, San José, CA 95112
| | - Bree Grillo-Hill
- Department of Biological Sciences, San José State University, San José, CA 95112
| |
Collapse
|
3
|
Pilesi E, Tesoriere G, Ferriero A, Mascolo E, Liguori F, Argirò L, Angioli C, Tramonti A, Contestabile R, Volontè C, Vernì F. Vitamin B6 deficiency cooperates with oncogenic Ras to induce malignant tumors in Drosophila. Cell Death Dis 2024; 15:388. [PMID: 38830901 PMCID: PMC11148137 DOI: 10.1038/s41419-024-06787-3] [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: 12/20/2023] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024]
Abstract
Vitamin B6 is a water-soluble vitamin which possesses antioxidant properties. Its catalytically active form, pyridoxal 5'-phosphate (PLP), is a crucial cofactor for DNA and amino acid metabolism. The inverse correlation between vitamin B6 and cancer risk has been observed in several studies, although dietary vitamin B6 intake sometimes failed to confirm this association. However, the molecular link between vitamin B6 and cancer remains elusive. Previous work has shown that vitamin B6 deficiency causes chromosome aberrations (CABs) in Drosophila and human cells, suggesting that genome instability may correlate the lack of this vitamin to cancer. Here we provide evidence in support of this hypothesis. Firstly, we show that PLP deficiency, induced by the PLP antagonists 4-deoxypyridoxine (4DP) or ginkgotoxin (GT), promoted tumorigenesis in eye larval discs transforming benign RasV12 tumors into aggressive forms. In contrast, PLP supplementation reduced the development of tumors. We also show that low PLP levels, induced by 4DP or by silencing the sgllPNPO gene involved in PLP biosynthesis, worsened the tumor phenotype in another Drosophila cancer model generated by concomitantly activating RasV12 and downregulating Discs-large (Dlg) gene. Moreover, we found that RasV12 eye discs from larvae reared on 4DP displayed CABs, reactive oxygen species (ROS) and low catalytic activity of serine hydroxymethyltransferase (SHMT), a PLP-dependent enzyme involved in thymidylate (dTMP) biosynthesis, in turn required for DNA replication and repair. Feeding RasV12 4DP-fed larvae with PLP or ascorbic acid (AA) plus dTMP, rescued both CABs and tumors. The same effect was produced by overexpressing catalase in RasV12 DlgRNAi 4DP-fed larvae, thus allowing to establish a relationship between PLP deficiency, CABs, and cancer. Overall, our data provide the first in vivo demonstration that PLP deficiency can impact on cancer by increasing genome instability, which is in turn mediated by ROS and reduced dTMP levels.
Collapse
Affiliation(s)
- Eleonora Pilesi
- Dept. of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Giulia Tesoriere
- Dept. of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Angelo Ferriero
- Dept. of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Elisa Mascolo
- Dept. of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Francesco Liguori
- Experimental Neuroscience and Neurological Disease Models, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- CNR, Institute for Systems Analysis and Computer Science, 00185, Rome, Italy
| | - Luca Argirò
- Dept. of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Chiara Angioli
- Dept. of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Angela Tramonti
- Institute of Molecular Biology and Pathology, 00185, Rome, Italy
| | - Roberto Contestabile
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza, University of Rome, 00185, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza, University of Rome, 00185, Rome, Italy
| | - Cinzia Volontè
- Experimental Neuroscience and Neurological Disease Models, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- CNR, Institute for Systems Analysis and Computer Science, 00185, Rome, Italy
| | - Fiammetta Vernì
- Dept. of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy.
| |
Collapse
|
4
|
Chen J, Zhao H, Liu M, Chen L. A new perspective on the autophagic and non-autophagic functions of the GABARAP protein family: a potential therapeutic target for human diseases. Mol Cell Biochem 2024; 479:1415-1441. [PMID: 37440122 DOI: 10.1007/s11010-023-04800-5] [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: 03/16/2023] [Accepted: 06/24/2023] [Indexed: 07/14/2023]
Abstract
Mammalian autophagy-related protein Atg8, including the LC3 subfamily and GABARAP subfamily. Atg8 proteins play a vital role in autophagy initiation, autophagosome formation and transport, and autophagy-lysosome fusion. GABARAP subfamily proteins (GABARAPs) share a high degree of homology with LC3 family proteins, and their unique roles are often overlooked. GABARAPs are as indispensable as LC3 in autophagy. Deletion of GABARAPs fails autophagy flux induction and autophagy lysosomal fusion, which leads to the failure of autophagy. GABARAPs are also involved in the transport of selective autophagy receptors. They are engaged in various particular autophagy processes, including mitochondrial autophagy, endoplasmic reticulum autophagy, Golgi autophagy, centrosome autophagy, and dorphagy. Furthermore, GABARAPs are closely related to the transport and delivery of the inhibitory neurotransmitter γ-GABAA and the angiotensin II AT1 receptor (AT1R), tumor growth, metastasis, and prognosis. GABARAPs also have been confirmed to be involved in various diseases, such as cancer, cardiovascular disease, and neurodegenerative diseases. In order to better understand the role and therapeutic potential of GABARAPs, this article comprehensively reviews the autophagic and non-autophagic functions of GABARAPs, as well as the research progress of the role and mechanism of GABARAPs in cancer, cardiovascular diseases and neurodegenerative diseases. It emphasizes the significance of GABARAPs in the clinical prevention and treatment of diseases, and may provide new therapeutic ideas and targets for human diseases. GABARAP and GABARAPL1 in the serum of cancer patients are positively correlated with the prognosis of patients, which can be used as a clinical biomarker, predictor and potential therapeutic target.
Collapse
Affiliation(s)
- Jiawei Chen
- Central Laboratory of Yan'nan Hospital Affiliated to Kunming, Medical University, Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, No. 245, Renmin East Road, Kunming, 650000, Yunnan, China
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hong Zhao
- Central Laboratory of Yan'nan Hospital Affiliated to Kunming, Medical University, Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, No. 245, Renmin East Road, Kunming, 650000, Yunnan, China
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Meiqing Liu
- Central Laboratory of Yan'nan Hospital Affiliated to Kunming, Medical University, Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, No. 245, Renmin East Road, Kunming, 650000, Yunnan, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
5
|
Rahmati S, Moeinafshar A, Rezaei N. The multifaceted role of extracellular vesicles (EVs) in colorectal cancer: metastasis, immune suppression, therapy resistance, and autophagy crosstalk. J Transl Med 2024; 22:452. [PMID: 38741166 DOI: 10.1186/s12967-024-05267-8] [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: 03/19/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer structures released by all cells and widely distributed in all biological fluids. EVs are implicated in diverse physiopathological processes by orchestrating cell-cell communication. Colorectal cancer (CRC) is one of the most common cancers worldwide, with metastasis being the leading cause of mortality in CRC patients. EVs contribute significantly to the advancement and spread of CRC by transferring their cargo, which includes lipids, proteins, RNAs, and DNAs, to neighboring or distant cells. Besides, they can serve as non-invasive diagnostic and prognostic biomarkers for early detection of CRC or be harnessed as effective carriers for delivering therapeutic agents. Autophagy is an essential cellular process that serves to remove damaged proteins and organelles by lysosomal degradation to maintain cellular homeostasis. Autophagy and EV release are coordinately activated in tumor cells and share common factors and regulatory mechanisms. Although the significance of autophagy and EVs in cancer is well established, the exact mechanism of their interplay in tumor development is obscure. This review focuses on examining the specific functions of EVs in various aspects of CRC, including progression, metastasis, immune regulation, and therapy resistance. Further, we overview emerging discoveries relevant to autophagy and EVs crosstalk in CRC.
Collapse
Affiliation(s)
- Soheil Rahmati
- Student Research Committee, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aysan Moeinafshar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Umargamwala R, Manning J, Dorstyn L, Denton D, Kumar S. Understanding Developmental Cell Death Using Drosophila as a Model System. Cells 2024; 13:347. [PMID: 38391960 PMCID: PMC10886741 DOI: 10.3390/cells13040347] [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: 01/23/2024] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Cell death plays an essential function in organismal development, wellbeing, and ageing. Many types of cell deaths have been described in the past 30 years. Among these, apoptosis remains the most conserved type of cell death in metazoans and the most common mechanism for deleting unwanted cells. Other types of cell deaths that often play roles in specific contexts or upon pathological insults can be classed under variant forms of cell death and programmed necrosis. Studies in Drosophila have contributed significantly to the understanding and regulation of apoptosis pathways. In addition to this, Drosophila has also served as an essential model to study the genetic basis of autophagy-dependent cell death (ADCD) and other relatively rare types of context-dependent cell deaths. Here, we summarise what is known about apoptosis, ADCD, and other context-specific variant cell death pathways in Drosophila, with a focus on developmental cell death.
Collapse
Affiliation(s)
- Ruchi Umargamwala
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Jantina Manning
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Loretta Dorstyn
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia; (J.M.); (L.D.)
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
7
|
Yang X, Xiong S, Zhao X, Jin J, Yang X, Du Y, Zhao L, He Z, Gong C, Guo L, Liang T. Orchestrating Cellular Balance: ncRNAs and RNA Interactions at the Dominant of Autophagy Regulation in Cancer. Int J Mol Sci 2024; 25:1561. [PMID: 38338839 PMCID: PMC10855840 DOI: 10.3390/ijms25031561] [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: 11/15/2023] [Revised: 12/15/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Autophagy, a complex and highly regulated cellular process, is critical for the maintenance of cellular homeostasis by lysosomal degradation of cellular debris, intracellular pathogens, and dysfunctional organelles. It has become an interesting and attractive topic in cancer because of its dual role as a tumor suppressor and cell survival mechanism. As a highly conserved pathway, autophagy is strictly regulated by diverse non-coding RNAs (ncRNAs), ranging from short and flexible miRNAs to lncRNAs and even circRNAs, which largely contribute to autophagy regulatory networks via complex RNA interactions. The potential roles of RNA interactions during autophagy, especially in cancer procession and further anticancer treatment, will aid our understanding of related RNAs in autophagy in tumorigenesis and cancer treatment. Herein, we mainly summarized autophagy-related mRNAs and ncRNAs, also providing RNA-RNA interactions and their potential roles in cancer prognosis, which may deepen our understanding of the relationships between various RNAs during autophagy and provide new insights into autophagy-related therapeutic strategies in personalized medicine.
Collapse
Affiliation(s)
- Xueni Yang
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Shizheng Xiong
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Xinmiao Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Jiaming Jin
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Xinbing Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.)
| | - Yajing Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.)
| | - Linjie Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Zhiheng He
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Chengjun Gong
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Li Guo
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (X.Y.); (S.X.); (X.Z.); (J.J.); (L.Z.); (Z.H.)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.)
| |
Collapse
|
8
|
Bakopoulos D, Golenkina S, Dark C, Christie EL, Sánchez-Sánchez BJ, Stramer BM, Cheng LY. Convergent insulin and TGF-β signalling drives cancer cachexia by promoting aberrant fat body ECM accumulation in a Drosophila tumour model. EMBO Rep 2023; 24:e57695. [PMID: 38014610 DOI: 10.15252/embr.202357695] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023] Open
Abstract
In this study, we found that in the adipose tissue of wildtype animals, insulin and TGF-β signalling converge via a BMP antagonist short gastrulation (sog) to regulate ECM remodelling. In tumour bearing animals, Sog also modulates TGF-β signalling to regulate ECM accumulation in the fat body. TGF-β signalling causes ECM retention in the fat body and subsequently depletes muscles of fat body-derived ECM proteins. Activation of insulin signalling, inhibition of TGF-β signalling, or modulation of ECM levels via SPARC, Rab10 or Collagen IV in the fat body, is able to rescue tissue wasting in the presence of tumour. Together, our study highlights the importance of adipose ECM remodelling in the context of cancer cachexia.
Collapse
Affiliation(s)
- Daniel Bakopoulos
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Callum Dark
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Brian M Stramer
- Kings College London, Randall Centre for Cell & Molecular Biophysics, London, UK
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
9
|
Sathiyaseelan P, Chittaranjan S, Kalloger SE, Chan J, Go NE, Jardon MA, Ho CJ, Hui T, Xu J, Chow C, Gao D, Johnson FD, Lockwood WW, Morin GB, Renouf DJ, Schaeffer DF, Gorski SM. Loss of ATG4B and ATG4A results in two-stage cell cycle defects in pancreatic ductal adenocarcinoma cells. J Cell Sci 2023; 136:jcs260644. [PMID: 37701987 PMCID: PMC10617609 DOI: 10.1242/jcs.260644] [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: 09/23/2022] [Accepted: 07/17/2023] [Indexed: 09/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits elevated levels of autophagy, which promote tumor progression and treatment resistance. ATG4B is an autophagy-related cysteine protease under consideration as a potential therapeutic target, but it is largely unexplored in PDAC. Here, we investigated the clinical and functional relevance of ATG4B expression in PDAC. Using two PDAC patient cohorts, we found that low ATG4B mRNA or protein expression is associated with worse patient survival outcomes, poorly differentiated PDAC tumors and a lack of survival benefit from adjuvant chemotherapy. In PDAC cell lines, ATG4B knockout reduced proliferation, abolished processing of LC3B (also known as MAP1LC3B), and reduced GABARAP and GABARAPL1 levels, but increased ATG4A levels. ATG4B and ATG4A double knockout lines displayed a further reduction in proliferation, characterized by delays in G1-S phase transition and mitosis. Pro-LC3B accumulated aberrantly at the centrosome with a concomitant increase in centrosomal proteins PCM1 and CEP131, which was rescued by exogenous ATG4B. The two-stage cell cycle defects following ATG4B and ATG4A loss have important therapeutic implications for PDAC.
Collapse
Affiliation(s)
- Paalini Sathiyaseelan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Suganthi Chittaranjan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Steve E. Kalloger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Pancreas Centre BC, Vancouver, BC, V5Z 1L8, Canada
| | - Jennifer Chan
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Nancy E. Go
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Mario A. Jardon
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Cally J. Ho
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Theodore Hui
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Jing Xu
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, Vancouver, BC, V6H 3Z6, Canada
| | - Dongxia Gao
- Genetic Pathology Evaluation Centre, Vancouver, BC, V6H 3Z6, Canada
| | - Fraser D. Johnson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Integrative Oncology, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - William W. Lockwood
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- Department of Integrative Oncology, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Gregg B. Morin
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6H 3N1, Canada
| | - Daniel J. Renouf
- Pancreas Centre BC, Vancouver, BC, V5Z 1L8, Canada
- Division of Medical Oncology, BC Cancer, Vancouver, BC, V5Z 4E6, Canada
| | - David F. Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
- Pancreas Centre BC, Vancouver, BC, V5Z 1L8, Canada
- Division of Anatomical Pathology, Vancouver General Hospital, Vancouver, BC, V5Z 1M9, Canada
| | - Sharon M. Gorski
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6H 3N1, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| |
Collapse
|
10
|
Ruze R, Song J, Yin X, Chen Y, Xu R, Wang C, Zhao Y. Mechanisms of obesity- and diabetes mellitus-related pancreatic carcinogenesis: a comprehensive and systematic review. Signal Transduct Target Ther 2023; 8:139. [PMID: 36964133 PMCID: PMC10039087 DOI: 10.1038/s41392-023-01376-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 03/26/2023] Open
Abstract
Research on obesity- and diabetes mellitus (DM)-related carcinogenesis has expanded exponentially since these two diseases were recognized as important risk factors for cancers. The growing interest in this area is prominently actuated by the increasing obesity and DM prevalence, which is partially responsible for the slight but constant increase in pancreatic cancer (PC) occurrence. PC is a highly lethal malignancy characterized by its insidious symptoms, delayed diagnosis, and devastating prognosis. The intricate process of obesity and DM promoting pancreatic carcinogenesis involves their local impact on the pancreas and concurrent whole-body systemic changes that are suitable for cancer initiation. The main mechanisms involved in this process include the excessive accumulation of various nutrients and metabolites promoting carcinogenesis directly while also aggravating mutagenic and carcinogenic metabolic disorders by affecting multiple pathways. Detrimental alterations in gastrointestinal and sex hormone levels and microbiome dysfunction further compromise immunometabolic regulation and contribute to the establishment of an immunosuppressive tumor microenvironment (TME) for carcinogenesis, which can be exacerbated by several crucial pathophysiological processes and TME components, such as autophagy, endoplasmic reticulum stress, oxidative stress, epithelial-mesenchymal transition, and exosome secretion. This review provides a comprehensive and critical analysis of the immunometabolic mechanisms of obesity- and DM-related pancreatic carcinogenesis and dissects how metabolic disorders impair anticancer immunity and influence pathophysiological processes to favor cancer initiation.
Collapse
Affiliation(s)
- Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Jianlu Song
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Chengcheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China.
| |
Collapse
|
11
|
Dutta SB, Linneweber GA, Andriatsilavo M, Hiesinger PR, Hassan BA. EGFR-dependent suppression of synaptic autophagy is required for neuronal circuit development. Curr Biol 2023; 33:517-532.e5. [PMID: 36640763 DOI: 10.1016/j.cub.2022.12.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/31/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023]
Abstract
The development of neuronal connectivity requires stabilization of dynamic axonal branches at sites of synapse formation. Models that explain how axonal branching is coupled to synaptogenesis postulate molecular regulators acting in a spatiotemporally restricted fashion to ensure branching toward future synaptic partners while also stabilizing the emerging synaptic contacts between such partners. We investigated this question using neuronal circuit development in the Drosophila brain as a model system. We report that epidermal growth factor receptor (EGFR) activity is required in presynaptic axonal branches during two distinct temporal intervals to regulate circuit wiring in the developing Drosophila visual system. EGFR is required early to regulate primary axonal branching. EGFR activity is then independently required at a later stage to prevent degradation of the synaptic active zone protein Bruchpilot (Brp). Inactivation of EGFR results in a local increase of autophagy in presynaptic branches and the translocation of active zone proteins into autophagic vesicles. The protection of synaptic material during this later interval of wiring ensures the stabilization of terminal branches, circuit connectivity, and appropriate visual behavior. Phenotypes of EGFR inactivation can be rescued by increasing Brp levels or downregulating autophagy. In summary, we identify a temporally restricted molecular mechanism required for coupling axonal branching and synaptic stabilization that contributes to the emergence of neuronal wiring specificity.
Collapse
Affiliation(s)
- Suchetana B Dutta
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié Salpêtrière, 75013 Paris, France; Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany; Einstein-BIH, Charité Universitätsmedizin, 10117 Berlin, Germany
| | | | - Maheva Andriatsilavo
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié Salpêtrière, 75013 Paris, France; Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany; Einstein-BIH, Charité Universitätsmedizin, 10117 Berlin, Germany
| | | | - Bassem A Hassan
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié Salpêtrière, 75013 Paris, France; Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany; Einstein-BIH, Charité Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
12
|
Ferreira A, Pereira F, Reis C, Oliveira MJ, Sousa MJ, Preto A. Crucial Role of Oncogenic KRAS Mutations in Apoptosis and Autophagy Regulation: Therapeutic Implications. Cells 2022; 11:cells11142183. [PMID: 35883626 PMCID: PMC9319879 DOI: 10.3390/cells11142183] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/05/2022] [Accepted: 07/10/2022] [Indexed: 11/16/2022] Open
Abstract
KRAS, one of the RAS protein family members, plays an important role in autophagy and apoptosis, through the regulation of several downstream effectors. In cancer cells, KRAS mutations confer the constitutive activation of this oncogene, stimulating cell proliferation, inducing autophagy, suppressing apoptosis, altering cell metabolism, changing cell motility and invasion and modulating the tumor microenvironment. In order to inhibit apoptosis, these oncogenic mutations were reported to upregulate anti-apoptotic proteins, including Bcl-xL and survivin, and to downregulate proteins related to apoptosis induction, including thymine-DNA glycosylase (TDG) and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL). In addition, KRAS mutations are known to induce autophagy in order to promote cell survival and tumor progression through MAPK and PI3K regulation. Thus, these mutations confer resistance to anti-cancer drug treatment and, consequently, result in poor prognosis. Several therapies have been developed in order to overcome KRAS-induced cell death resistance and the downstream signaling pathways blockade, especially by combining MAPK and PI3K inhibitors, which demonstrated promising results. Understanding the involvement of KRAS mutations in apoptosis and autophagy regulation, might bring new avenues to the discovery of therapeutic approaches for CRCs harboring KRAS mutations.
Collapse
Affiliation(s)
- Anabela Ferreira
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (A.F.); (F.P.); (M.J.S.)
- Institute of Science and Innovation for Bio-Sustainability (IB-S), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Flávia Pereira
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (A.F.); (F.P.); (M.J.S.)
- Institute of Science and Innovation for Bio-Sustainability (IB-S), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (C.R.); (M.J.O.)
- Institute of Biomedical Engineering (INEB), University of Porto, 4200-135 Porto, Portugal
| | - Celso Reis
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (C.R.); (M.J.O.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - Maria José Oliveira
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (C.R.); (M.J.O.)
- Institute of Biomedical Engineering (INEB), University of Porto, 4200-135 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Maria João Sousa
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (A.F.); (F.P.); (M.J.S.)
- Institute of Science and Innovation for Bio-Sustainability (IB-S), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Ana Preto
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (A.F.); (F.P.); (M.J.S.)
- Institute of Science and Innovation for Bio-Sustainability (IB-S), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- Correspondence: ; Tel.: +351-253-601524
| |
Collapse
|
13
|
Chen Z, Lee HJ, Kim H, Cho S, Kim K. δ-Catenin promotes cell migration and invasion via Bcl-2-regulated suppression of autophagy in prostate cancer cells. Am J Cancer Res 2022; 12:108-122. [PMID: 35141007 PMCID: PMC8822292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/08/2021] [Indexed: 06/14/2023] Open
Abstract
As a member of the catenin family, δ-catenin is overexpressed in many cancers, including prostate cancer, and the role of δ-catenin in prostate tumor growth has been reported. However, the involvement of δ-catenin in the migration and invasion of prostate cancer has rarely been studied. In this study, we innovatively proposed that δ-catenin would enhance the migration and invasion ability of prostate cancer cells. It is worth noting that the molecular mechanism underlying the effect involved the downregulation of autophagy. We demonstrated that δ-catenin could suppress autophagy by Bcl-2-regulated disruption of the Beclin1-Vps34 autophagosome complex. Furthermore, the effect of δ-catenin on promoting cell migration and invasion was dependent upon β-catenin-mediated Bcl-2 transcription. Finally, using rapamycin and bafilomycin, we largely confirmed that the degradation of Snails by autolysosomes may be related to δ-catenin regulated migration and invasion. Overall, our results indicated that δ-catenin promoted cell migration and invasion of prostate cancer cells via Bcl-2-regulated autophagy suppression.
Collapse
Affiliation(s)
- Zhiwei Chen
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National UniversityGwangju 61186, Korea
- School of Pharmaceutical Science, Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Hyoung Jae Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National UniversityGwangju 61186, Korea
| | - Hangun Kim
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National UniversitySunchon 57922, Korea
| | - Sayeon Cho
- College of Pharmacy, Chung-Ang UniversitySeoul 06974, Korea
| | - Kwonseop Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National UniversityGwangju 61186, Korea
| |
Collapse
|
14
|
A Blueprint for Cancer-Related Inflammation and Host Innate Immunity. Cells 2021; 10:cells10113211. [PMID: 34831432 PMCID: PMC8623541 DOI: 10.3390/cells10113211] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/27/2021] [Accepted: 11/10/2021] [Indexed: 12/30/2022] Open
Abstract
Both in situ and allograft models of cancer in juvenile and adult Drosophila melanogaster fruit flies offer a powerful means for unravelling cancer gene networks and cancer-host interactions. They can also be used as tools for cost-effective drug discovery and repurposing. Moreover, in situ modeling of emerging tumors makes it possible to address cancer initiating events-a black box in cancer research, tackle the innate antitumor immune responses to incipient preneoplastic cells and recurrent growing tumors, and decipher the initiation and evolution of inflammation. These studies in Drosophila melanogaster can serve as a blueprint for studies in more complex organisms and help in the design of mechanism-based therapies for the individualized treatment of cancer diseases in humans. This review focuses on new discoveries in Drosophila related to the diverse innate immune responses to cancer-related inflammation and the systemic effects that are so detrimental to the host.
Collapse
|
15
|
Tumor-derived MMPs regulate cachexia in a Drosophila cancer model. Dev Cell 2021; 56:2664-2680.e6. [PMID: 34473940 DOI: 10.1016/j.devcel.2021.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 07/09/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022]
Abstract
Cachexia, the wasting syndrome commonly observed in advanced cancer patients, accounts for up to one-third of cancer-related mortalities. We have established a Drosophila larval model of organ wasting whereby epithelial overgrowth in eye-antennal discs leads to wasting of the adipose tissue and muscles. The wasting is associated with fat-body remodeling and muscle detachment and is dependent on tumor-secreted matrix metalloproteinase 1 (Mmp1). Mmp1 can both modulate TGFβ signaling in the fat body and disrupt basement membrane (BM)/extracellular matrix (ECM) protein localization in both the fat body and the muscle. Inhibition of TGFβ signaling or Mmps in the fat body/muscle using a QF2-QUAS binary expression system rescues muscle wasting in the presence of tumor. Altogether, our study proposes that tumor-derived Mmps are central mediators of organ wasting in cancer cachexia.
Collapse
|
16
|
Wang K, Shao X, Tian Z, Liu L, Zhang C, Tan C, Zhang J, Ling P, Liu F, Chen Q, Diao J, Mao Z. A Continuous Add-On Probe Reveals the Nonlinear Enlargement of Mitochondria in Light-Activated Oncosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004566. [PMID: 34197052 PMCID: PMC8425930 DOI: 10.1002/advs.202004566] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/03/2021] [Indexed: 05/25/2023]
Abstract
Oncosis, depending on DNA damage and mitochondrial swelling, is an important approach for treating cancer and other diseases. However, little is known about the behavior of mitochondria during oncosis, due to the lack of probes for in situ visual illumination of the mitochondrial membrane and mtDNA. Herein, a mitochondrial lipid and mtDNA dual-labeled probe, MitoMN, and a continuous add-on assay, are designed to image the dynamic process of mitochondria in conditions that are unobservable with current mitochondrial probes. Meanwhile, the MitoMN can induce oncosis in a light-activated manner, which results in the enlargement of mitochondria and the death of cancer cells. Using structured illumination microscopy (SIM), MitoMN-stained mitochondria with a dual-color response reveals, for the first time, how swelled mitochondria interacts and fuses with each other for a nonlinear enlargement to accelerate oncosis into an irreversible stage. With this sign of irreversible oncosis revealed by MitoMN, oncosis can be segregated into three stages, including before oncosis, initial oncosis, and accelerated oncosis.
Collapse
Affiliation(s)
- Kang‐Nan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Xintian Shao
- Institute of Materia MedicaShandong First Medical University & Shandong Academy of Medical SciencesJinan250000P. R. China
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
| | - Zhiqi Tian
- Department of Molecular Genetics, Biochemistry, and MicrobiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
| | - Liu‐Yi Liu
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Chengying Zhang
- Institute of Materia MedicaShandong First Medical University & Shandong Academy of Medical SciencesJinan250000P. R. China
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
- School of Pharmaceutical SciencesShandong UniversityJinan250101P. R. China
| | - Cai‐Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| | - Jie Zhang
- Advanced Medical Research Institute/Translational Medicine Core Facility of Advanced Medical Research InstituteShandong UniversityJinan250101P. R. China
| | - Peixue Ling
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
- School of Pharmaceutical SciencesShandong UniversityJinan250101P. R. China
| | - Fei Liu
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
- School of Pharmaceutical SciencesShandong UniversityJinan250101P. R. China
| | - Qixin Chen
- Institute of Materia MedicaShandong First Medical University & Shandong Academy of Medical SciencesJinan250000P. R. China
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
- Shandong Academy of Pharmaceutical ScienceKey Laboratory of BiopharmaceuticalsEngineering Laboratory of Polysaccharide DrugsNational‐Local Joint Engineering Laboratory of Polysaccharide DrugsJinan250101P. R. China
| | - Jiajie Diao
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnati45267USA
| | - Zong‐Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic ChemistrySchool of ChemistryState Key Laboratory of Oncology in South ChinaSun Yat‐Sen UniversityGuangzhou510275P. R. China
| |
Collapse
|
17
|
Guo C, Zhao Y. Autophagy in pancreatic cancer. J Mol Cell Biol 2021; 13:786-790. [PMID: 34459894 PMCID: PMC8782586 DOI: 10.1093/jmcb/mjab053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Congting Guo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ying Zhao
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
18
|
Zada S, Hwang JS, Ahmed M, Lai TH, Pham TM, Elashkar O, Kim DR. Cross talk between autophagy and oncogenic signaling pathways and implications for cancer therapy. Biochim Biophys Acta Rev Cancer 2021; 1876:188565. [PMID: 33992723 DOI: 10.1016/j.bbcan.2021.188565] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
Autophagy is a highly conserved metabolic process involved in the degradation of intracellular components including proteins and organelles. Consequently, it plays a critical role in recycling metabolic energy for the maintenance of cellular homeostasis in response to various stressors. In cancer, autophagy either suppresses or promotes cancer progression depending on the stage and cancer type. Epithelial-mesenchymal transition (EMT) and cancer metastasis are directly mediated by oncogenic signal proteins including SNAI1, SLUG, ZEB1/2, and NOTCH1, which are functionally correlated with autophagy. In this report, we discuss the crosstalk between oncogenic signaling pathways and autophagy followed by possible strategies for cancer treatment via regulation of autophagy. Although autophagy affects EMT and cancer metastasis, the overall signaling pathways connecting cancer progression and autophagy are still illusive. In general, autophagy plays a critical role in cancer cell survival by providing a minimum level of energy via self-digestion. Thus, cancer cells face nutrient limitations and challenges under stress during EMT and metastasis. Conversely, autophagy acts as a potential cancer suppressor by degrading oncogenic proteins, which are essential for cancer progression, and by removing damaged components such as mitochondria to enhance genomic stability. Therefore, autophagy activators or inhibitors represent possible cancer therapeutics. We further discuss the regulation of autophagy-dependent degradation of oncogenic proteins and its functional correlation with oncogenic signaling pathways, with potential applications in cancer therapy.
Collapse
Affiliation(s)
- Sahib Zada
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Trang Minh Pham
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Omar Elashkar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea.
| |
Collapse
|
19
|
Glaucocalyxin A induces apoptosis and autophagy in tongue squamous cell carcinoma cells by regulating ROS. Cancer Chemother Pharmacol 2021; 88:235-246. [PMID: 33904969 DOI: 10.1007/s00280-021-04285-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/17/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Tongue squamous cell carcinoma (TSCC) is the most common highly invasive oral cancer. Glaucocalyxin A (GLA) is a diterpenoid component isolated from Rabdosia japonica var. with anti-bacterial and anti-cancer biological properties. However, the role of GLA in human TSCC remains uncertain. The aim of this paper was to investigate the anti-cancer effect of GLA on TSCC cells as well as its underlying mechanism. METHODS Cell viability and growth were analyzed by CCK-8 assay and colony formation, respectively. DAPI staining and flow cytometry assay were used to detect the cell apoptosis. Lysotracker Red staining was used to observe the lysosomes and autolysosomes of TSCC cells. ROS fluorescent probe was used to test the intracellular ROS levels. Western blotting was used to detect the expression levels of apoptosis- and autophagy-related proteins. RESULTS GLA inhibits the cell viability and growth in TSCC cells. GLA induces TSCC cells apoptosis, autophagy and ROS production in a time- and concentration-dependent manner. In addition, GLA inhibits the viability of TSCC cells by inducing intracellular ROS production. Finally, GLA triggers ROS-dependent apoptosis and autophagy in TSCC cells. CONCLUSION Our results consistently suggested that GLA can induce apoptosis and autophagy in TSCC cells by generating ROS. GLA may serve as a promising therapeutic drug for overcoming TSCC.
Collapse
|
20
|
Alvarez-Ochoa E, Froldi F, Cheng LY. Interorgan communication in development and cancer. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e394. [PMID: 32852143 DOI: 10.1002/wdev.394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 06/22/2020] [Accepted: 07/16/2020] [Indexed: 11/10/2022]
Abstract
Studies in model organisms have demonstrated that extensive communication occurs between distant organs both during development and in diseases such as cancer. Organs communicate with each other to coordinate growth and reach the correct size, while the fate of tumor cells depend on the outcome of their interaction with the immune system and peripheral tissues. In this review, we outline recent studies in Drosophila, which have enabled an improved understanding of the complex crosstalk between organs in the context of both organismal and tumor growth. We argue that Drosophila is a powerful model organism for studying these interactions, and these studies have the potential for improving our understanding of signaling pathways and candidate factors that mediate this conserved interorgan crosstalk. This article is categorized under: Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Early Embryonic Development > Development to the Basic Body Plan Invertebrate Organogenesis > Flies.
Collapse
Affiliation(s)
- Edel Alvarez-Ochoa
- Peter MacCallum Cancer Centre, Parkville, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Francesca Froldi
- Peter MacCallum Cancer Centre, Parkville, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Parkville, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,The Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
21
|
Zhang F, Pirooznia M, Xu H. Mitochondria regulate intestinal stem cell proliferation and epithelial homeostasis through FOXO. Mol Biol Cell 2020; 31:1538-1549. [PMID: 32374658 PMCID: PMC7359575 DOI: 10.1091/mbc.e19-10-0560] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A metabolic transition from glycolysis to oxidative phosphorylation is often associated with differentiation of many types of stem cells. However, the link between mitochondrial respiration and stem cells' behavior is not fully understood. We genetically disrupted electron transport chain (ETC) complexes in the intestinal stem cells (ISCs) of Drosophila. We found that ISCs carrying impaired ETC proliferated much more slowly than normal and produced very few enteroblasts, which failed to further differentiate into enterocytes. One of the main impediments to ISC proliferation and lineage specification appeared to be abnormally elevated forkhead box O (FOXO) signaling in the ETC-deficient ISCs, as genetically suppressing the signaling pathway partially restored the number of enterocytes. Contrary to common belief, reactive oxygen species (ROS) accumulation did not appear to mediate the ETC mutant phenotype. Our results demonstrate that mitochondrial respiration is essential for Drosophila ISC proliferation and lineage specification in vivo and acts at least partially by repressing endogenous FOXO signaling.
Collapse
Affiliation(s)
- Fan Zhang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Mehdi Pirooznia
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Hong Xu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
22
|
Drosophila as a model to understand autophagy deregulation in human disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020. [PMID: 32620249 DOI: 10.1016/bs.pmbts.2020.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Autophagy has important functions in normal physiology to maintain homeostasis and protect against cellular stresses by the removal of harmful cargos such as dysfunctional organelles, protein aggregates and invading pathogens. The deregulation of autophagy is a hallmark of many diseases and therapeutic targeting of autophagy is highly topical. With the complex role of autophagy in disease it is essential to understand the genetic and molecular basis of the contribution of autophagy to pathogenesis. The model organism, Drosophila, provides a genetically amenable system to dissect out the contribution of autophagy to human disease models. Here we review the roles of autophagy in human disease and how autophagy studies in Drosophila have contributed to the understanding of pathophysiology.
Collapse
|
23
|
Lin L, Xia L, Tang D, Dai Y, Chen W. Analysis of autophagy-related genes and associated noncoding RNAs and transcription factors in digestive system tumors. Future Oncol 2019; 15:4141-4154. [PMID: 31802711 DOI: 10.2217/fon-2019-0341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To investigate the autophagy-related gene (ATG) expression and the associated noncoding RNAs (ncRNA) and transcription factors (TF) in digestive system tumors (DST). Methods: We systematically investigated the ATG expression in DST by weighted gene correlation network analysis, crosstalk connection, functional analysis and Pivot analysis. Results: ATGs were clustered into six modules with co-expression in DST. Functional analysis revealed that six ATG-related modules were enriched in biological pathways involved in tumorigenesis. Pivot analysis identified key ncRNAs and TFs, which are essential for the pathogenesis, clinical diagnosis and treatment of DST. Conclusion: Our study highlights the crucial roles of ncRNA and TFs in the identification of potential biomarkers or therapeutic targets for DST.
Collapse
Affiliation(s)
- Liewen Lin
- Department of Gastrointestinal Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science & Technology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, PR China
| | - Ligang Xia
- Department of Gastrointestinal Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science & Technology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, PR China
| | - Donge Tang
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science & Technology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, PR China
| | - Yong Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science & Technology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, PR China
| | - Wenbiao Chen
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Disease, Collaborative Innovation Center for Diagnosis & Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science & Technology, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, PR China
| |
Collapse
|
24
|
Bhattacharjee A, Szabó Á, Csizmadia T, Laczkó-Dobos H, Juhász G. Understanding the importance of autophagy in human diseases using Drosophila. J Genet Genomics 2019; 46:157-169. [PMID: 31080044 DOI: 10.1016/j.jgg.2019.03.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/19/2022]
Abstract
Autophagy is a lysosome-dependent intracellular degradation pathway that has been implicated in the pathogenesis of various human diseases, either positively or negatively impacting disease outcomes depending on the specific context. The majority of medical conditions including cancer, neurodegenerative diseases, infections and immune system disorders and inflammatory bowel disease could probably benefit from therapeutic modulation of the autophagy machinery. Drosophila represents an excellent model animal to study disease mechanisms thanks to its sophisticated genetic toolkit, and the conservation of human disease genes and autophagic processes. Here, we provide an overview of the various autophagy pathways observed both in flies and human cells (macroautophagy, microautophagy and chaperone-mediated autophagy), and discuss Drosophila models of the above-mentioned diseases where fly research has already helped to understand how defects in autophagy genes and pathways contribute to the relevant pathomechanisms.
Collapse
Affiliation(s)
- Arindam Bhattacharjee
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári Krt. 62., Szeged, H-6726, Hungary
| | - Áron Szabó
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári Krt. 62., Szeged, H-6726, Hungary
| | - Tamás Csizmadia
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Pázmány Sétány 1/C, Budapest, H-1117, Hungary
| | - Hajnalka Laczkó-Dobos
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári Krt. 62., Szeged, H-6726, Hungary
| | - Gábor Juhász
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári Krt. 62., Szeged, H-6726, Hungary; Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Pázmány Sétány 1/C, Budapest, H-1117, Hungary.
| |
Collapse
|
25
|
Khezri R, Rusten TE. Autophagy and Tumorigenesis in Drosophila. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1167:113-127. [PMID: 31520352 DOI: 10.1007/978-3-030-23629-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The resurgence of Drosophila as a recognized model for carcinogenesis has contributed greatly to our conceptual advance and mechanistic understanding of tumor growth in vivo. With its powerful genetics, Drosophila has emerged as a prime model organism to study cell biology and physiological functions of autophagy. This has enabled exploration of the contributions of autophagy in several tumor models. Here we review the literature of autophagy related to tumorigenesis in Drosophila. Functional analysis of core autophagy components does not provide proof for a classical tumor suppression role for autophagy alone. Autophagy both serve to suppress or support tumor growth. These effects are context-specific, depending on cell type and oncogenic or tumor suppressive lesion. Future delineation of how autophagy impinges on tumorigenesis will demand to untangle in detail, the regulation and flux of autophagy in the respective tumor models. The downstream tumor-regulative roles of autophagy through organelle homeostasis, metabolism, selective autophagy or alternative mechanisms remain largely unexplored.
Collapse
Affiliation(s)
- Rojyar Khezri
- Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, The Medical Faculty, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Tor Erik Rusten
- Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, The Medical Faculty, University of Oslo, Oslo, Norway.
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
26
|
Zoranovic T, Manent J, Willoughby L, Matos de Simoes R, La Marca JE, Golenkina S, Cuiping X, Gruber S, Angjeli B, Kanitz EE, Cronin SJF, Neely GG, Wernitznig A, Humbert PO, Simpson KJ, Mitsiades CS, Richardson HE, Penninger JM. A genome-wide Drosophila epithelial tumorigenesis screen identifies Tetraspanin 29Fb as an evolutionarily conserved suppressor of Ras-driven cancer. PLoS Genet 2018; 14:e1007688. [PMID: 30325918 PMCID: PMC6203380 DOI: 10.1371/journal.pgen.1007688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 10/26/2018] [Accepted: 09/11/2018] [Indexed: 12/15/2022] Open
Abstract
Oncogenic mutations in the small GTPase Ras contribute to ~30% of human cancers. However, Ras mutations alone are insufficient for tumorigenesis, therefore it is paramount to identify cooperating cancer-relevant signaling pathways. We devised an in vivo near genome-wide, functional screen in Drosophila and discovered multiple novel, evolutionarily-conserved pathways controlling Ras-driven epithelial tumorigenesis. Human gene orthologs of the fly hits were significantly downregulated in thousands of primary tumors, revealing novel prognostic markers for human epithelial tumors. Of the top 100 candidate tumor suppressor genes, 80 were validated in secondary Drosophila assays, identifying many known cancer genes and multiple novel candidate genes that cooperate with Ras-driven tumorigenesis. Low expression of the confirmed hits significantly correlated with the KRASG12 mutation status and poor prognosis in pancreatic cancer. Among the novel top 80 candidate cancer genes, we mechanistically characterized the function of the top hit, the Tetraspanin family member Tsp29Fb, revealing that Tsp29Fb regulates EGFR signaling, epithelial architecture and restrains tumor growth and invasion. Our functional Drosophila screen uncovers multiple novel and evolutionarily conserved epithelial cancer genes, and experimentally confirmed Tsp29Fb as a key regulator of EGFR/Ras induced epithelial tumor growth and invasion. Cancer involves the cooperative interaction of many gene mutations. The Ras signaling pathway is upregulated in many human cancers, but upregulated Ras signaling alone is not sufficient to induce malignant tumors. We have undertaken a genome-wide genetic screen using a transgenic RNAi library in the vinegar fly, Drosophila melanogaster, to identify tumor suppressor genes that cooperate with the Ras oncogene (RasV12) in conferring overgrown invasive tumors. We stratified the hits by analyzing the expression of human orthologs of these genes in human epithelial cancers, revealing genes that were strongly downregulated in human cancer. By conducting secondary genetic interaction tests, we validated 80 of the top 100 genes. Pathway analysis of these genes revealed that 55 fell into known pathways involved in human cancer, whereas 25 were unique genes. We then confirmed the tumor suppressor properties of one of these genes, Tsp29Fb, encoding a Tetraspanin membrane protein, and showed that Tsp29Fb functions as a tumor suppressor by inhibiting Ras signaling and by maintaining epithelial cell polarity. Altogether, our study has revealed novel Ras-cooperating tumor suppressors in Drosophila and suggests that these genes may also be involved in human cancer.
Collapse
Affiliation(s)
- Tamara Zoranovic
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Jan Manent
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Lee Willoughby
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ricardo Matos de Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - John E. La Marca
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Sofya Golenkina
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Xia Cuiping
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Susanne Gruber
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Belinda Angjeli
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Elisabeth Eva Kanitz
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - Shane J. F. Cronin
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
| | - G. Gregory Neely
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
- The Charles Perkins Centre, School of Life & Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Patrick O. Humbert
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, and Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kaylene J. Simpson
- Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, and Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
- Victorian Center for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Constantine S. Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Helena E. Richardson
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, and Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
- * E-mail: (HER); (JMP)
| | - Josef M. Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Science, Campus Vienna BioCentre, Vienna, Austria
- * E-mail: (HER); (JMP)
| |
Collapse
|
27
|
Benhra N, Barrio L, Muzzopappa M, Milán M. Chromosomal Instability Induces Cellular Invasion in Epithelial Tissues. Dev Cell 2018; 47:161-174.e4. [DOI: 10.1016/j.devcel.2018.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/19/2018] [Accepted: 08/22/2018] [Indexed: 01/08/2023]
|
28
|
Bishop E, Bradshaw TD. Autophagy modulation: a prudent approach in cancer treatment? Cancer Chemother Pharmacol 2018; 82:913-922. [PMID: 30182146 PMCID: PMC6267659 DOI: 10.1007/s00280-018-3669-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/12/2018] [Indexed: 01/07/2023]
Abstract
Autophagy is a tightly controlled process comprising lysosomal degradation and recycling of cellular proteins and organelles. In cancer, its paradoxical dual role of cytoprotection and cytotoxicity is context-dependent and controversial. Autophagy primarily acts as a mechanism of tumour suppression, by maintenance of genomic integrity and prevention of proliferation and inflammation. This, combined with immune-surveillance capabilities and autophagy's implicated role in cell death, acts to prevent tumour initiation. However, established tumours exploit autophagy to survive cellular stresses in the hostile tumour microenvironment. This can lead to therapy resistance, one of the biggest challenges facing current anti-cancer approaches. Autophagy modulation is an exciting area of clinical development, attempting to harness this fundamental process as an anti-cancer strategy. Autophagy induction could potentially prevent tumour formation and enhance anti-cancer immune responses. In addition, drug-induced autophagy could be used to kill cancer cells, particularly those in which the apoptotic machinery is defective. Conversely, autophagy inhibition may help to sensitise resistant cancer cells to conventional chemotherapies and specifically target autophagy-addicted tumours. Currently, hydroxychloroquine is in phase I and II clinical trials in combination with several standard chemotherapies, whereas direct, deliberate autophagy induction remains to be tested clinically. More comprehensive understanding of the roles of autophagy throughout different stages of carcinogenesis has potential to guide development of novel therapeutic strategies to eradicate cancer cells.
Collapse
Affiliation(s)
- Eleanor Bishop
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Tracey D Bradshaw
- School of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| |
Collapse
|
29
|
Parvy JP, Hodgson JA, Cordero JB. Drosophila as a Model System to Study Nonautonomous Mechanisms Affecting Tumour Growth and Cell Death. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7152962. [PMID: 29725601 PMCID: PMC5872677 DOI: 10.1155/2018/7152962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/04/2018] [Indexed: 12/26/2022]
Abstract
The study of cancer has represented a central focus in medical research for over a century. The great complexity and constant evolution of the pathology require the use of multiple research model systems and interdisciplinary approaches. This is necessary in order to achieve a comprehensive understanding into the mechanisms driving disease initiation and progression, to aid the development of appropriate therapies. In recent decades, the fruit fly Drosophila melanogaster and its associated powerful genetic tools have become a very attractive model system to study tumour-intrinsic and non-tumour-derived processes that mediate tumour development in vivo. In this review, we will summarize recent work on Drosophila as a model system to study cancer biology. We will focus on the interactions between tumours and their microenvironment, including extrinsic mechanisms affecting tumour growth and how tumours impact systemic host physiology.
Collapse
Affiliation(s)
- Jean-Philippe Parvy
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Joseph A. Hodgson
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Julia B. Cordero
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
30
|
Modelling Cooperative Tumorigenesis in Drosophila. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4258387. [PMID: 29693007 PMCID: PMC5859872 DOI: 10.1155/2018/4258387] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/21/2018] [Indexed: 12/13/2022]
Abstract
The development of human metastatic cancer is a multistep process, involving the acquisition of several genetic mutations, tumour heterogeneity, and interactions with the surrounding microenvironment. Due to the complexity of cancer development in mammals, simpler model organisms, such as the vinegar fly, Drosophila melanogaster, are being utilized to provide novel insights into the molecular mechanisms involved. In this review, we highlight recent advances in modelling tumorigenesis using the Drosophila model, focusing on the cooperation of oncogenes or tumour suppressors, and the interaction of mutant cells with the surrounding tissue in epithelial tumour initiation and progression.
Collapse
|
31
|
Pérez E, Lindblad JL, Bergmann A. Tumor-promoting function of apoptotic caspases by an amplification loop involving ROS, macrophages and JNK in Drosophila. eLife 2017; 6:e26747. [PMID: 28853394 PMCID: PMC5779227 DOI: 10.7554/elife.26747] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/28/2017] [Indexed: 01/04/2023] Open
Abstract
Apoptosis and its molecular mediators, the caspases, have long been regarded as tumor suppressors and one hallmark of cancer is 'Evading Apoptosis'. However, recent work has suggested that apoptotic caspases can also promote proliferation and tumor growth under certain conditions. How caspases promote proliferation and how cells are protected from the potentially harmful action of apoptotic caspases is largely unknown. Here, we show that although caspases are activated in a well-studied neoplastic tumor model in Drosophila, oncogenic mutations of the proto-oncogene Ras (RasV12) maintain tumorous cells in an 'undead'-like condition and transform caspases from tumor suppressors into tumor promotors. Instead of killing cells, caspases now promote the generation of intra- and extracellular reactive oxygen species (ROS). One function of the ROS is the recruitment and activation of macrophage-like immune cells which in turn signal back to tumorous epithelial cells to activate oncogenic JNK signaling. JNK further promotes and amplifies caspase activity, thereby constituting a feedback amplification loop. Interfering with the amplification loop strongly reduces the neoplastic behavior of these cells and significantly improves organismal survival. In conclusion, RasV12-modified caspases initiate a feedback amplification loop involving tumorous epithelial cells and macrophage-like immune cells that is necessary for uncontrolled tumor growth and invasive behavior.
Collapse
Affiliation(s)
- Ernesto Pérez
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Jillian L Lindblad
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Andreas Bergmann
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| |
Collapse
|