1
|
Chen Y, Li Q, Wang Z, Sun LV, Hou SX. A novel NFKB1 agonist remodels tumor microenvironment and activates dendritic cells to promote anti-tumor immunity in colorectal cancer. J Transl Med 2025; 23:561. [PMID: 40394677 PMCID: PMC12090520 DOI: 10.1186/s12967-025-06576-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND The immunosuppressive nature of the tumor microenvironment (TME) and the existence of cancer stem cells (CSCs) present significant hurdles in tumor therapy. The identification of therapeutic agents that can target both CSCs and the TME could be a potential approach to overcome treatment resistance. METHODS We conducted an in vivo chemical screen to identify F1929-1458, which is capable of eliciting an organism-wide response to destroy stem cell tumors in Drosophila. We then performed functional validation using a mouse colorectal cancer graft tumor model established with the CT26 cell line characterized by its high content of CSCs. Single-cell sequencing was employed to analyze alterations in the TME. Small molecule pull-down mass spectrometry, cellular thermal shift assay, drug affinity experiment, and molecular docking were utilized to identify the target of F1929-1458. An in vitro co-culture system was applied to establish that the damage-associated molecular patterns (DAMPs) released by the tumor cells are accountable for the activation of dendritic cells (DCs). RESULTS We demonstrated that F1929-1458 treatment enhanced T cell infiltration and T cell mediated tumor regression, its anti-tumor effect was nullified in nude mice and was abolished after anti-CD3 neutralizing antibody treatment. We found that F1929-1458 binds NFKB1 to activate the NF-κB signaling pathway in tumor cells. The activation further elicits cellular stress, causing tumor cells to release DAMPs (HMGB1-gDNA complex, ATP, and OxLDL). These DAMPs, in turn, stimulate the cGAS-STING and NLRP3 inflammasome pathways in DCs, resulting in the generation of type I IFNs and IL-1β. These cytokines facilitate the maturation of DCs and antigen presentation, ultimately enhancing T cell-mediated anti-tumor immunity. Additionally, we showed that the combination of F1929-1458 and the anti-PD-1 antibody exhibited a synergistic anti-tumor effect. CONCLUSION Our study identified a novel NFKB1 agonist that promotes anti-tumor immunity by remodeling the TME and activating DCs and that may provide a new way to overcome resistance to current anti-tumor immunotherapy in colorectal cancer.
Collapse
Affiliation(s)
- Ying Chen
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Children's Hospital, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Qiaoming Li
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Children's Hospital, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Zixiang Wang
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Children's Hospital, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Ling V Sun
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Children's Hospital, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| | - Steven X Hou
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Children's Hospital, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
2
|
Zheng C, Zheng J, Wang X, Zhang Y, Ma X, He L. Two-pore-domain potassium channel Sandman regulates intestinal stem cell homeostasis and tumorigenesis in Drosophila melanogaster. J Genet Genomics 2025:S1673-8527(25)00147-X. [PMID: 40381822 DOI: 10.1016/j.jgg.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Potassium channels regulate diverse biological processes, ranging from cell proliferation to immune responses. However, the functions of potassium homeostasis and its regulatory mechanisms in adult stem cells and tumors remain poorly characterized. Here, we identify Sandman, a two-pore-domain potassium channel in Drosophila, as an essential regulator for the proliferation of intestinal stem cells and malignant tumors, while dispensable for the normal development processes. Mechanistically, loss of sandman elevates intracellular K+ concentration, leading to growth inhibition. This phenotype is rescued by pharmacological reduction of intracellular K+ levels using the K+ ionophore. Conversely, overexpression of sandman triggers stem cell death in most regions of the midgut, inhibits tumor growth, and induces a Notch loss-of-function phenotype in the posterior midgut. These effects are mediated predominantly via the induction of endoplasmic reticulum (ER) stress, as demonstrated by the complete rescue of phenotypes through the co-expression of Ire1 or Xbp1s. Additionally, human homologs of Sandman demonstrated similar ER stress-inducing capabilities, suggesting an evolutionarily conserved relationship between this channel and ER stress. Together, our findings identify Sandman as a shared regulatory node that governs Drosophila adult stem cell dynamics and tumorigenesis through bioelectric homeostasis, and reveal a link between the two-pore potassium channel and ER stress signaling.
Collapse
Affiliation(s)
- Chen Zheng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jiadong Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Xin Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yue Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xianjue Ma
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China.
| | - Li He
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
3
|
Aloisi M, Deloriea J, Casey C, Pino A, Morciano P, Grifoni D, Gamberi C. DROSOPHILA: THE CENTURY-LONG FLIGHT FROM THE WILD TO THE PATIENT. MEDICAL SCIENCE PULSE 2025; 19:1-15. [PMID: 40276781 PMCID: PMC12021435 DOI: 10.5604/01.3001.0054.9627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Abstract
Background: Evolutionary conservation of key biological pathways between the fruit fly Drosophila melanogaster and humans and reduced genetic redundancy have long made flies a valuable genetic model organism. Thanks to the arsenal of sophisticated genetic tools developed and refined by the fly community, the use of Drosophila has expanded well beyond basic research. From the fundamental notion that genes are located on chromosomes to modeling human complex diseases such as cancer and neurological disorders, to designing fly "avatar" lines that precisely reproduce the specific mutations found in single cancer patients for personalized medicine, Drosophila continues to fuel biomedical advances. Numerous examples of drug testing in flies have yielded novel drug candidates, new uses for approved drugs, and applications for rapid drug optimization in modern approaches combining biology with medicinal chemistry. Thanks to the effectiveness of "fly pharmacology" approaches, Drosophila is also proficiently used to study the mechanism of action of environmental pollutants that represent a serious concern to human health. This review traces the history of some of the main advances in Drosophila biomedical and cancer research.
Collapse
Affiliation(s)
- Massimo Aloisi
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
- Department of Life, Health and Environmental Sciences, L'Aquila University, Italy
| | - Jay Deloriea
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| | - Cody Casey
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| | - Alexia Pino
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| | - Patrizia Morciano
- Department of Life, Health and Environmental Sciences, L'Aquila University, Italy
- INFN-Laboratori Nazionali del Gran Sasso, Assergi, L'Aquila, Italy
| | - Daniela Grifoni
- Department of Life, Health and Environmental Sciences, L'Aquila University, Italy
| | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| |
Collapse
|
4
|
Song H, Kim S, Han JE, Kang KH, Koh H. PDH Inhibition in Drosophila Ameliorates Sensory Dysfunction Induced by Vincristine Treatment in the Chemotherapy-Induced Peripheral Neuropathy Models. Biomedicines 2025; 13:783. [PMID: 40299339 PMCID: PMC12025153 DOI: 10.3390/biomedicines13040783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Chemotherapy-induced peripheral neuropathy (CIPN) is a significant dose-limiting side effect of many effective anticancer agents, including vincristine. While CIPN adversely affects both oncological outcomes and the quality of life for cancer patients, the in vivo mechanisms behind CIPN pathology remain largely unknown, and effective treatments have yet to be developed. In this study, we established a novel Drosophila model of CIPN using vincristine to explore the molecular mechanisms underlying this condition. Methods: We assessed the impact of vincristine exposure on thermal nociception in Drosophila larvae using a programmable heat probe. Additionally, we investigated vincristine-induced mitochondrial dysfunction and dendritic abnormalities in class IV dendritic arborization (C4da) neurons with various fluorescent protein markers. Results: We found a dose-dependent increase in thermal hypersensitivity, accompanied by changes in the sensory dendrites of C4da neurons in vincristine-treated fly larvae. Moreover, vincristine significantly enhanced mitochondrial ROS production and mitophagy-a selective autophagy that targets dysfunctional mitochondria-indicating vincristine-induced mitochondrial dysfunction within C4da neurons. Surprisingly, inhibiting the pyruvate dehydrogenase complex (PDH), a key mitochondrial metabolic enzyme complex, effectively rescued the mitochondrial and sensory abnormalities caused by vincristine. Conclusions: Findings from this first Drosophila model of vincristine-induced peripheral neuropathy (VIPN) suggest that mitochondrial dysfunction plays a critical role in VIPN pathology, representing PDH as a potential target for the treatment of VIPN.
Collapse
Affiliation(s)
- Harim Song
- Department of Pharmacology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (H.S.); (S.K.); (J.E.H.)
- Department of Translational Biomedical Sciences, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Sohee Kim
- Department of Pharmacology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (H.S.); (S.K.); (J.E.H.)
- Department of Translational Biomedical Sciences, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Ji Eun Han
- Department of Pharmacology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (H.S.); (S.K.); (J.E.H.)
- Department of Translational Biomedical Sciences, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Kyong-hwa Kang
- Department of Pharmacology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (H.S.); (S.K.); (J.E.H.)
- Neuroscience Translational Research Solution Center, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Hyongjong Koh
- Department of Pharmacology, Dong-A University College of Medicine, Busan 49201, Republic of Korea; (H.S.); (S.K.); (J.E.H.)
- Department of Translational Biomedical Sciences, Dong-A University College of Medicine, Busan 49201, Republic of Korea
- Neuroscience Translational Research Solution Center, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| |
Collapse
|
5
|
Diaz JEL, Barcessat V, Bahamon C, Hecht C, Das TK, Cagan RL. Functional exploration of copy number alterations in a Drosophila model of triple-negative breast cancer. Dis Model Mech 2024; 17:dmm050191. [PMID: 38721669 PMCID: PMC11247506 DOI: 10.1242/dmm.050191] [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: 03/14/2023] [Accepted: 04/30/2024] [Indexed: 07/04/2024] Open
Abstract
Accounting for 10-20% of breast cancer cases, triple-negative breast cancer (TNBC) is associated with a disproportionate number of breast cancer deaths. One challenge in studying TNBC is its genomic profile: with the exception of TP53 loss, most breast cancer tumors are characterized by a high number of copy number alterations (CNAs), making modeling the disease in whole animals challenging. We computationally analyzed 186 CNA regions previously identified in breast cancer tumors to rank genes within each region by likelihood of acting as a tumor driver. We then used a Drosophila p53-Myc TNBC model to identify 48 genes as functional drivers. To demonstrate the utility of this functional database, we established six 3-hit models; altering candidate genes led to increased aspects of transformation as well as resistance to the chemotherapeutic drug fluorouracil. Our work provides a functional database of CNA-associated TNBC drivers, and a template for an integrated computational/whole-animal approach to identify functional drivers of transformation and drug resistance within CNAs in other tumor types.
Collapse
Affiliation(s)
- Jennifer E L Diaz
- Department of Cell, Development, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Internal Medicine, UCLA David Geffen School of Medicine, CA 90095, USA
| | - Vanessa Barcessat
- Department of Cell, Development, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christian Bahamon
- Department of Cell, Development, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chana Hecht
- Department of Cell, Development, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tirtha K Das
- Department of Cell, Development, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ross L Cagan
- Department of Cell, Development, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- School of Cancer Sciences and Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow G61 1BD, UK
| |
Collapse
|
6
|
Pranoto IKA, Kwon YV. Protocol to analyze Drosophila intestinal tumor cellular heterogeneity using immunofluorescence imaging and nuclear size quantification. STAR Protoc 2024; 5:102946. [PMID: 38470911 PMCID: PMC10945268 DOI: 10.1016/j.xpro.2024.102946] [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: 12/07/2023] [Revised: 01/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Drosophila intestinal tumors show an extended cellular heterogeneity. We devise a protocol to assess tumor cell heterogeneity by employing nuclear size measurement and immunofluorescence-based cell lineage analysis. We describe steps for intestinal dissection, staining, and imaging, followed by detailed procedures for nuclear size analysis. This approach detects overall heterogeneity across the entire tumor cell population and deviations within specific cell populations. The procedure is also applicable for analyzing the heterogeneity of wild-type intestinal cells in various contexts. For complete details on the use and execution of this protocol, please refer to Pranoto et al.1.
Collapse
Affiliation(s)
| | - Young V Kwon
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
7
|
Leung NY, Xu C, Li JSS, Ganguly A, Meyerhof GT, Regimbald-Dumas Y, Lane EA, Breault DT, He X, Perrimon N, Montell C. Gut tumors in flies alter the taste valence of an anti-tumorigenic bitter compound. Curr Biol 2024; 34:2623-2632.e5. [PMID: 38823383 PMCID: PMC11308992 DOI: 10.1016/j.cub.2024.04.082] [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: 08/25/2022] [Revised: 03/28/2024] [Accepted: 04/30/2024] [Indexed: 06/03/2024]
Abstract
The sense of taste is essential for survival, as it allows animals to distinguish between foods that are nutritious from those that are toxic. However, innate responses to different tastants can be modulated or even reversed under pathological conditions. Here, we examined whether and how the internal status of an animal impacts taste valence by using Drosophila models of hyperproliferation in the gut. In all three models where we expressed proliferation-inducing transgenes in intestinal stem cells (ISCs), hyperproliferation of ISCs caused a tumor-like phenotype in the gut. While tumor-bearing flies had no deficiency in overall food intake, strikingly, they exhibited an increased gustatory preference for aristolochic acid (ARI), which is a bitter and normally aversive plant-derived chemical. ARI had anti-tumor effects in all three of our gut hyperproliferation models. For other aversive chemicals we tested that are bitter but do not have anti-tumor effects, gut tumors did not affect avoidance behaviors. We demonstrated that bitter-sensing gustatory receptor neurons (GRNs) in tumor-bearing flies respond normally to ARI. Therefore, the internal pathology of gut hyperproliferation affects neural circuits that determine taste valence postsynaptic to GRNs rather than altering taste identity by GRNs. Overall, our data suggest that increased consumption of ARI may represent an attempt at self-medication. Finally, although ARI's potential use as a chemotherapeutic agent is limited by its known toxicity in the liver and kidney, our findings suggest that tumor-bearing flies might be a useful animal model to screen for novel anti-tumor drugs.
Collapse
Affiliation(s)
- Nicole Y Leung
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Chiwei Xu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Joshua Shing Shun Li
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Anindya Ganguly
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Geoff T Meyerhof
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Yannik Regimbald-Dumas
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth A Lane
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Xi He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Craig Montell
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
8
|
Kurogi Y, Mizuno Y, Kamiyama T, Niwa R. The intestinal stem cell/enteroblast-GAL4 driver, escargot-GAL4, also manipulates gene expression in the juvenile hormone-synthesizing organ of Drosophila melanogaster. Sci Rep 2024; 14:9631. [PMID: 38671036 PMCID: PMC11053112 DOI: 10.1038/s41598-024-60269-2] [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: 01/12/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
Intestinal stem cells (ISCs) of the fruit fly, Drosophila melanogaster, offer an excellent genetic model to explore homeostatic roles of ISCs in animal physiology. Among available genetic tools, the escargot (esg)-GAL4 driver, expressing the yeast transcription factor gene, GAL4, under control of the esg gene promoter, has contributed significantly to ISC studies. This driver facilitates activation of genes of interest in proximity to a GAL4-binding element, Upstream Activating Sequence, in ISCs and progenitor enteroblasts (EBs). While esg-GAL4 has been considered an ISC/EB-specific driver, recent studies have shown that esg-GAL4 is also active in other tissues, such as neurons and ovaries. Therefore, the ISC/EB specificity of esg-GAL4 is questionable. In this study, we reveal esg-GAL4 expression in the corpus allatum (CA), responsible for juvenile hormone (JH) production. When driving the oncogenic gene, RasV12, esg-GAL4 induces overgrowth in ISCs/EBs as reported, but also increases CA cell number and size. Consistent with this observation, animals alter expression of JH-response genes. Our data show that esg-GAL4-driven gene manipulation can systemically influence JH-mediated animal physiology, arguing for cautious use of esg-GAL4 as a "specific" ISC/EB driver to examine ISC/EB-mediated animal physiology.
Collapse
Affiliation(s)
- Yoshitomo Kurogi
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Yosuke Mizuno
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Takumi Kamiyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
9
|
Sharmin Z, Samarah H, Aldaya Bourricaudy R, Ochoa L, Serbus LR. Cross-validation of chemical and genetic disruption approaches to inform host cellular effects on Wolbachia abundance in Drosophila. Front Microbiol 2024; 15:1364009. [PMID: 38591028 PMCID: PMC10999648 DOI: 10.3389/fmicb.2024.1364009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction Endosymbiotic Wolbachia bacteria are widespread in nature, present in half of all insect species. The success of Wolbachia is supported by a commensal lifestyle. Unlike bacterial pathogens that overreplicate and harm host cells, Wolbachia infections have a relatively innocuous intracellular lifestyle. This raises important questions about how Wolbachia infection is regulated. Little is known about how Wolbachia abundance is controlled at an organismal scale. Methods This study demonstrates methodology for rigorous identification of cellular processes that affect whole-body Wolbachia abundance, as indicated by absolute counts of the Wolbachia surface protein (wsp) gene. Results Candidate pathways, associated with well-described infection scenarios, were identified. Wolbachia-infected fruit flies were exposed to small molecule inhibitors known for targeting those same pathways. Sequential tests in D. melanogaster and D. simulans yielded a subset of chemical inhibitors that significantly affected whole-body Wolbachia abundance, including the Wnt pathway disruptor, IWR-1 and the mTOR pathway inhibitor, Rapamycin. The implicated pathways were genetically retested for effects in D. melanogaster, using inducible RNAi expression driven by constitutive as well as chemically-induced somatic GAL4 expression. Genetic disruptions of armadillo, tor, and ATG6 significantly affected whole-body Wolbachia abundance. Discussion As such, the data corroborate reagent targeting and pathway relevance to whole-body Wolbachia infection. The results also implicate Wnt and mTOR regulation of autophagy as important for regulation of Wolbachia titer.
Collapse
Affiliation(s)
- Zinat Sharmin
- Department of Biological Sciences, Florida International University, Miami, FL, United States
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Hani Samarah
- Department of Biological Sciences, Florida International University, Miami, FL, United States
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Rafael Aldaya Bourricaudy
- Department of Biological Sciences, Florida International University, Miami, FL, United States
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Laura Ochoa
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, United States
| | - Laura Renee Serbus
- Department of Biological Sciences, Florida International University, Miami, FL, United States
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, United States
| |
Collapse
|
10
|
Pfefferkorn RM, Mortzfeld BM, Fink C, von Frieling J, Bossen J, Esser D, Kaleta C, Rosenstiel P, Heine H, Roeder T. Recurrent Phases of Strict Protein Limitation Inhibit Tumor Growth and Restore Lifespan in A Drosophila Intestinal Cancer Model. Aging Dis 2024; 15:226-244. [PMID: 37962464 PMCID: PMC10796089 DOI: 10.14336/ad.2023.0517] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/17/2023] [Indexed: 11/15/2023] Open
Abstract
Diets that restrict caloric or protein intake offer a variety of benefits, including decreasing the incidence of cancer. However, whether such diets pose a substantial therapeutic benefit as auxiliary cancer treatments remains unclear. We determined the effects of severe protein depletion on tumorigenesis in a Drosophila melanogaster intestinal tumor model, using a human RAF gain-of-function allele. Severe and continuous protein restriction significantly reduced tumor growth but resulted in premature death. Therefore, we developed a diet in which short periods of severe protein restriction alternated cyclically with periods of complete feeding. This nutritional regime reduced tumor mass, restored gut functionality, and rescued the lifespan of oncogene-expressing flies to the levels observed in healthy flies on a continuous, fully nutritious diet. Furthermore, this diet reduced the chemotherapy-induced stem cell activity associated with tumor recurrence. Transcriptome analysis revealed long-lasting changes in the expression of key genes involved in multiple major developmental signaling pathways. Overall, the data suggest that recurrent severe protein depletion effectively mimics the health benefits of continuous protein restriction, without undesired nutritional shortcomings. This provides seminal insights into the mechanisms of the memory effect required to maintain the positive effects of protein restriction throughout the phases of a full diet. Finally, the repetitive form of strict protein restriction is an ideal strategy for adjuvant cancer therapy that is useful in many tumor contexts.
Collapse
Affiliation(s)
- Roxana M. Pfefferkorn
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Benedikt M. Mortzfeld
- Department of Cell and Developmental Biology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Christine Fink
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Jakob von Frieling
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Judith Bossen
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| | - Daniela Esser
- Department of Neuroimmunology, Institute of Clinical Chemistry, University Medical Center Schleswig-Holstein, Kiel, Germany.
| | - Christoph Kaleta
- Department Medical Systems Biology, Institute for Experimental Medicine, Kiel University, Germany.
| | - Philip Rosenstiel
- Department Molecular Cell Biology, Institute for Clinical Molecular Biology, Kiel University, Germany.
| | - Holger Heine
- Division of Innate Immunity, Research Center Borstel - Leibniz Lung Center, Borstel, Germany.
| | - Thomas Roeder
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.
| |
Collapse
|
11
|
Pranoto IKA, Lee J, Kwon YV. The roles of the native cell differentiation program aberrantly recapitulated in Drosophila intestinal tumors. Cell Rep 2023; 42:113245. [PMID: 37837622 PMCID: PMC10872463 DOI: 10.1016/j.celrep.2023.113245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/11/2023] [Accepted: 09/26/2023] [Indexed: 10/16/2023] Open
Abstract
Many tumors recapitulate the developmental and differentiation program of their tissue of origin, a basis for tumor cell heterogeneity. Although stem-cell-like tumor cells are well studied, the roles of tumor cells undergoing differentiation remain to be elucidated. We employ Drosophila genetics to demonstrate that the differentiation program of intestinal stem cells is crucial for enabling intestinal tumors to invade and induce non-tumor-autonomous phenotypes. The differentiation program that generates absorptive cells is aberrantly recapitulated in the intestinal tumors generated by activation of the Yap1 ortholog Yorkie. Inhibiting it allows stem-cell-like tumor cells to grow but suppresses invasiveness and reshapes various phenotypes associated with cachexia-like wasting by altering the expression of tumor-derived factors. Our study provides insight into how a native differentiation program determines a tumor's capacity to induce advanced cancer phenotypes and suggests that manipulating the differentiation programs co-opted in tumors might alleviate complications of cancer, including cachexia.
Collapse
Affiliation(s)
| | - Jiae Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Young V Kwon
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
12
|
Zhou J, Boutros M. Intestinal stem cells and their niches in homeostasis and disease. Cells Dev 2023; 175:203862. [PMID: 37271243 DOI: 10.1016/j.cdev.2023.203862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/21/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Tissues such as the intestine harbor stem cells that have remarkable functional plasticity in response to a dynamic environment. To adapt to the environment, stem cells constantly receive information from their surrounding microenvironment (also called the 'niche') that instructs them how to adapt to changes. The Drosophila midgut shows morphological and functional similarities to the mammalian small intestine and has been a useful model system to study signaling events in stem cells and tissue homeostasis. In this review, we summarize the current understanding of the Drosophila midgut regarding how stem cells communicate with microenvironmental niches including enteroblasts, enterocytes, enteroendocrine cells and visceral muscles to coordinate tissue regeneration and homeostasis. In addition, distant cells such as hemocytes or tracheal cells have been shown to interact with stem cells and influence the development of intestinal diseases. We discuss the contribution of stem cell niches in driving or counteracting disease progression, and review conceptual advances derived from the Drosophila intestine as a model for stem cell biology.
Collapse
Affiliation(s)
- Jun Zhou
- German Cancer Research Center (DKFZ), Heidelberg University, Division Signaling and Functional Genomics, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany; School of Biomedical Sciences, Hunan University, Changsha, China.
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Heidelberg University, Division Signaling and Functional Genomics, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany.
| |
Collapse
|
13
|
Voutyraki C, Choromidis A, Meligkounaki A, Vlachopoulos NA, Theodorou V, Grammenoudi S, Athanasiadis E, Monticelli S, Giangrande A, Delidakis C, Zacharioudaki E. Growth deregulation and interaction with host hemocytes contribute to tumor progression in a Drosophila brain tumor model. Proc Natl Acad Sci U S A 2023; 120:e2221601120. [PMID: 37549261 PMCID: PMC10438840 DOI: 10.1073/pnas.2221601120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/27/2023] [Indexed: 08/09/2023] Open
Abstract
Tumors constantly interact with their microenvironment. Here, we present data on a Notch-induced neural stem cell (NSC) tumor in Drosophila, which can be immortalized by serial transplantation in adult hosts. This tumor arises in the larva by virtue of the ability of Notch to suppress early differentiation-promoting factors in NSC progeny. Guided by transcriptome data, we have addressed both tumor-intrinsic and microenvironment-specific factors and how they contribute to tumor growth and host demise. The growth promoting factors Myc, Imp, and Insulin receptor in the tumor cells are important for tumor expansion and killing of the host. From the host's side, hemocytes, professional phagocytic blood cells, are found associated with tumor cells. Phagocytic receptors, like NimC1, are needed in hemocytes to enable them to capture and engulf tumor cells, restricting their growth. In addition to their protective role, hemocytes may also increase the host's morbidity by their propensity to produce damaging extracellular reactive oxygen species.
Collapse
Affiliation(s)
- Chrysanthi Voutyraki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Alexandros Choromidis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Anastasia Meligkounaki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Nikolaos Andreas Vlachopoulos
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Vasiliki Theodorou
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
| | - Sofia Grammenoudi
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672Athens, Greece
| | - Emmanouil Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens, 11527Athens, Greece
- Medical Image and Signal Processing Laboratory, Department of Biomedical Engineering, University of West Attica, 12243Athens, Greece
| | - Sara Monticelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400Strasbourg, France
- Centre National de la Recherche Scientifique, UMR7104Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1258Strasbourg, France
- Université de Strasbourg, 67404Strasbourg, France
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400Strasbourg, France
- Centre National de la Recherche Scientifique, UMR7104Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1258Strasbourg, France
- Université de Strasbourg, 67404Strasbourg, France
| | - Christos Delidakis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Evanthia Zacharioudaki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
| |
Collapse
|
14
|
Zhang Y, Chen R, Gong L, Huang W, Li P, Zhai Z, Ling E. Regulation of intestinal stem cell activity by a mitotic cell cycle regulator Polo in Drosophila. G3 (BETHESDA, MD.) 2023; 13:jkad084. [PMID: 37154439 PMCID: PMC10234410 DOI: 10.1093/g3journal/jkad084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/31/2023] [Indexed: 05/10/2023]
Abstract
Maintaining a definite and stable pool of dividing stem cells plays an important role in organ development. This process requires an appropriate progression of mitosis for proper spindle orientation and polarity to ensure the ability of stem cells to proliferate and differentiate correctly. Polo-like kinases (Plks)/Polo are the highly conserved serine/threonine kinases involved in the initiation of mitosis as well as in the progression of the cell cycle. Although numerous studies have investigated the mitotic defects upon loss of Plks/Polo in cells, little is known about the in vivo consequences of stem cells with abnormal Polo activity in the context of tissue and organism development. The current study aimed to investigate this question using the Drosophila intestine, an organ dynamically maintained by the intestinal stem cells (ISCs). The results indicated that the polo depletion caused a reduction in the gut size due to a gradual decrease in the number of functional ISCs. Interestingly, the polo-deficient ISCs showed an extended G2/M phase and aneuploidy and were subsequently eliminated by premature differentiation into enterocytes (ECs). In contrast, the constitutively active Polo (poloT182D) suppressed ISC proliferation, induced abnormal accumulation of β-tubulin in cells, and drove ISC loss via apoptosis. Therefore, Polo activity should be properly maintained for optimal stem cell function. Further analysis suggested that polo was a direct target gene of Sox21a, a Sox transcription factor that critically regulates stem cell activity. Together, this study provided a novel perspective on the correlation between the progression of mitosis and the ISC function in Drosophila.
Collapse
Affiliation(s)
- Ying Zhang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Rongbing Chen
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Liyuan Gong
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wuren Huang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
| | - Ping Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zongzhao Zhai
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Erjun Ling
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
| |
Collapse
|
15
|
Okada M, Takano T, Ikegawa Y, Ciesielski H, Nishida H, Yoo SK. Oncogenic stress-induced Netrin is a humoral signaling molecule that reprograms systemic metabolism in Drosophila. EMBO J 2023:e111383. [PMID: 37140455 DOI: 10.15252/embj.2022111383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/01/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
Cancer exerts pleiotropic, systemic effects on organisms, leading to health deterioration and eventually to organismal death. How cancer induces systemic effects on remote organs and the organism itself still remains elusive. Here we describe a role for NetrinB (NetB), a protein with a particularly well-characterized role as a tissue-level axon guidance cue, in mediating oncogenic stress-induced organismal, metabolic reprogramming as a systemic humoral factor. In Drosophila, Ras-induced dysplastic cells upregulate and secrete NetB. Inhibition of either NetB from the transformed tissue or its receptor in the fat body suppresses oncogenic stress-induced organismal death. NetB from the dysplastic tissue remotely suppresses carnitine biosynthesis in the fat body, which is critical for acetyl-CoA generation and systemic metabolism. Supplementation of carnitine or acetyl-CoA ameliorates organismal health under oncogenic stress. This is the first identification, to our knowledge, of a role for the Netrin molecule, which has been studied extensively for its role within tissues, in humorally mediating systemic effects of local oncogenic stress on remote organs and organismal metabolism.
Collapse
Affiliation(s)
- Morihiro Okada
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
| | - Tomomi Takano
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
| | - Yuko Ikegawa
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hanna Ciesielski
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Division of Developmental Biology and Regenerative Medicine, Kobe University, Kobe, Japan
| | - Hiroshi Nishida
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Division of Cell Physiology, Kobe University, Kobe, Japan
| | - Sa Kan Yoo
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
- Division of Developmental Biology and Regenerative Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
16
|
Jaime MDLA, Salem GH, Martinez DJ, Karott S, Flores A, Palmer CD, Mahadevaraju S, Krynitsky J, Garmendia-Cedillos M, Anderson S, Harbison S, Pohida TJ, Ludington WB, Oliver B. Whole Animal Feeding FLat (WAFFL): a complete and comprehensive validation of a novel, high-throughput fly experimentation system. G3 (BETHESDA, MD.) 2023; 13:6989864. [PMID: 36650008 PMCID: PMC9997563 DOI: 10.1093/g3journal/jkad012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/08/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
Non-mammalian model organisms have been essential for our understanding of the mechanisms that control development, disease, and physiology, but they are underutilized in pharmacological and toxicological phenotypic screening assays due to their low throughput in comparison with cell-based screens. To increase the utility of using Drosophila melanogaster in screening, we designed the Whole Animal Feeding FLat (WAFFL), a novel, flexible, and complete system for feeding, monitoring, and assaying flies in a high-throughput format. Our 3D printed system is compatible with inexpensive and readily available, commercial 96-well plate consumables and equipment. Experimenters can change the diet at will during the experiment and video record for behavior analysis, enabling precise dosing, measurement of feeding, and analysis of behavior in a 96-well plate format.
Collapse
Affiliation(s)
- Maria D L A Jaime
- Department of Embryology, Carnegie Institution of Washington, Baltimore, MD 21218, USA.,Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Ghadi H Salem
- Instrument Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 12 South Dr, Bethesda, MD 20892, USA
| | - Daniel J Martinez
- Department of Embryology, Carnegie Institution of Washington, Baltimore, MD 21218, USA
| | - Sean Karott
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Alejandra Flores
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA.,Department of Physiology and Biophysics, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Cameron D Palmer
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Sharvani Mahadevaraju
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA.,Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| | - Jonathan Krynitsky
- Instrument Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 12 South Dr, Bethesda, MD 20892, USA
| | - Marcial Garmendia-Cedillos
- Instrument Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 12 South Dr, Bethesda, MD 20892, USA
| | - Sarah Anderson
- Instrument Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 12 South Dr, Bethesda, MD 20892, USA
| | - Susan Harbison
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20814, USA
| | - Thomas J Pohida
- Instrument Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 12 South Dr, Bethesda, MD 20892, USA
| | - William B Ludington
- Department of Embryology, Carnegie Institution of Washington, Baltimore, MD 21218, USA.,Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brian Oliver
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA.,Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA
| |
Collapse
|
17
|
Moskal N, Visanji NP, Gorbenko O, Narasimhan V, Tyrrell H, Nash J, Lewis PN, McQuibban GA. An AI-guided screen identifies probucol as an enhancer of mitophagy through modulation of lipid droplets. PLoS Biol 2023; 21:e3001977. [PMID: 36862640 PMCID: PMC9980794 DOI: 10.1371/journal.pbio.3001977] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/22/2022] [Indexed: 03/03/2023] Open
Abstract
Failures in mitophagy, a process by which damaged mitochondria are cleared, results in neurodegeneration, while enhancing mitophagy promotes the survival of dopaminergic neurons. Using an artificial intelligence platform, we employed a natural language processing approach to evaluate the semantic similarity of candidate molecules to a set of well-established mitophagy enhancers. Top candidates were screened in a cell-based mitochondrial clearance assay. Probucol, a lipid-lowering drug, was validated across several orthogonal mitophagy assays. In vivo, probucol improved survival, locomotor function, and dopaminergic neuron loss in zebrafish and fly models of mitochondrial damage. Probucol functioned independently of PINK1/Parkin, but its effects on mitophagy and in vivo depended on ABCA1, which negatively regulated mitophagy following mitochondrial damage. Autophagosome and lysosomal markers were elevated by probucol treatment in addition to increased contact between lipid droplets (LDs) and mitochondria. Conversely, LD expansion, which occurs following mitochondrial damage, was suppressed by probucol and probucol-mediated mitophagy enhancement required LDs. Probucol-mediated LD dynamics changes may prime the cell for a more efficient mitophagic response to mitochondrial damage.
Collapse
Affiliation(s)
- Natalia Moskal
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Naomi P. Visanji
- Edmund J Safra Program in Parkinson’s Disease and Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, Canada
| | - Olena Gorbenko
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Vijay Narasimhan
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital and Department of Medicine and Physiology, University of Toronto, Toronto, Canada
| | - Hannah Tyrrell
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Jess Nash
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Peter N. Lewis
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | | |
Collapse
|
18
|
Zhai J, Li W, Liu X, Wang D, Zhang D, Liu Y, Liang X, Chen Z. Tiny Drosophila intestinal stem cells, big power. Cell Biol Int 2022; 47:3-14. [PMID: 36177490 DOI: 10.1002/cbin.11911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/12/2022] [Accepted: 09/12/2022] [Indexed: 11/12/2022]
Abstract
The signaling pathways are highly conserved between Drosophila and mammals concerning intestinal development, regeneration, and disease. The powerful genetic tools of Drosophila make it a valuable and convenient alternative to answer basic biological questions that can not be addressed using mammalian models. In this review, we discuss recent advances in how we use fly midgut to answer the following key questions: (1) How intestine stem cell niches are established; (2) which factors control asymmetric division of stem cells; (3) how intestinal cells interact with environmental factors, such as tissue damage, microbiota, and diet; (4) how to screen aging/cancer-related factors or drugs by fly intestine stem cells.
Collapse
Affiliation(s)
- Jingbo Zhai
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Wanyang Li
- Medical College, Inner Mongolia Minzu University, Tongliao, China
| | - Xin Liu
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Di Wang
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Dongli Zhang
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| | - Yanli Liu
- Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, China
| | - Xiuwen Liang
- Hulunbuir City People's Hospital, Hulunbuir City, China
| | - Zeliang Chen
- Medical College, Inner Mongolia Minzu University, Tongliao, China.,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao, China.,Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, China
| |
Collapse
|
19
|
Weina T, Ying L, Yiwen W, Huan-Huan Q. What we have learnt from Drosophila model organism: the coordination between insulin signaling pathway and tumor cells. Heliyon 2022; 8:e09957. [PMID: 35874083 PMCID: PMC9304707 DOI: 10.1016/j.heliyon.2022.e09957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/25/2022] [Accepted: 07/11/2022] [Indexed: 02/08/2023] Open
Abstract
Cancer development is related to a variety of signaling pathways which mediate various cellular processes including growth, survival, division and competition of cells, as well as cell-cell interaction. The insulin signaling pathway interacts with different pathways and plays a core role in the regulations of all these processes. In this study, we reviewed recent studies on the relationship between the insulin signaling pathway and tumors using the Drosophila melanogaster model. We found that on one hand, the insulin pathway is normally hyperactive in tumor cells, which promotes tumor growth, and on the other hand, tumor cells can suppress the growth of healthy tissues via inhibition of their insulin pathway. Moreover, systematic disruption in glucose homeostasis also facilitates cancer development by different mechanisms. The studies on how the insulin network regulates the behaviors of cancer cells may help to discover new therapeutic treatments for cancer.
Collapse
Affiliation(s)
- Tang Weina
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Li Ying
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Wang Yiwen
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Qiao Huan-Huan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, Tianjin, China
| |
Collapse
|
20
|
Zhu JY, Huang X, Fu Y, Wang Y, Zheng P, Liu Y, Han Z. Pharmacological or genetic inhibition of hypoxia signaling attenuates oncogenic RAS-induced cancer phenotypes. Dis Model Mech 2022; 15:272327. [PMID: 34580712 PMCID: PMC8617310 DOI: 10.1242/dmm.048953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/18/2021] [Indexed: 12/30/2022] Open
Abstract
Oncogenic Ras mutations are highly prevalent in hematopoietic malignancies. However, it is difficult to directly target oncogenic RAS proteins for therapeutic intervention. We have developed a Drosophila acute myeloid leukemia model induced by human KRASG12V, which exhibits a dramatic increase in myeloid-like leukemia cells. We performed both genetic and drug screens using this model. The genetic screen identified 24 candidate genes able to attenuate the oncogenic RAS-induced phenotype, including two key hypoxia pathway genes HIF1A and ARNT (HIF1B). The drug screen revealed that echinomycin, an inhibitor of HIF1A, can effectively attenuate the leukemia phenotype caused by KRASG12V. Furthermore, we showed that echinomycin treatment can effectively suppress oncogenic RAS-driven leukemia cell proliferation, using both human leukemia cell lines and a mouse xenograft model. These data suggest that inhibiting the hypoxia pathway could be an effective treatment approach and that echinomycin is a promising targeted drug to attenuate oncogenic RAS-induced cancer phenotypes. This article has an associated First Person interview with the first author of the paper. Summary: Hypoxia pathway inhibition, either genetically or pharmacologically, rescues RAS-induced oncogenesis in a Drosophila acute myeloid leukemia model, mouse xenograft model and human leukemia cells.
Collapse
Affiliation(s)
- Jun-Yi Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Division of Immunotherapy, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xiaohu Huang
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Division of Immunotherapy, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yulong Fu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yin Wang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pan Zheng
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yang Liu
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Division of Immunotherapy, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
21
|
Brás R, Monteiro A, Sunkel CE, Resende LP. Aneuploidy facilitates dysplastic and tumorigenic phenotypes in the Drosophila gut. Biol Open 2021; 10:bio058623. [PMID: 33948620 PMCID: PMC8576263 DOI: 10.1242/bio.058623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
Aneuploidy has been strongly linked to cancer development, and published evidence has suggested that aneuploidy can have an oncogenic or a tumor suppressor role depending on the tissue context. Using the Drosophila midgut as a model, we have recently described that adult intestinal stem cells (ISCs), do not activate programmed cell death upon aneuploidy induction, leading to an increase in ISC proliferation rate, and tissue dysplasia. How aneuploidy impacts ISCs in intestinal tumorigenic models remains to be investigated, and it represents a very important biological question to address since data from multiple in vivo models suggests that the cellular impact of aneuploidy is highly dependent on the cellular and tissue context. Using manipulation of different genetic pathways such as EGFR, JAK-STAT and Notch that cause dysplastic phenotypes in the Drosophila gut, we found that concomitant aneuploidy induction by impairment of the spindle assembly checkpoint (SAC) consistently leads to a more severe progression of intestinal dysplasia or tumorigenesis. This is characterized by an accumulation of progenitor cells, high tissue cell density and higher stem cell proliferation rates, revealing an additive or synergistic effect depending on the misregulated pathway in which aneuploidy was induced. Thus, our data suggests that in the Drosophila gut, both dysplasia and tumorigenic phenotypes can be fueled by inducing genomic instability of resident stem cells.
Collapse
Affiliation(s)
- Rita Brás
- Instituto de Investigaçaõ e Inovaçaõ em Saúde, Universidade do Porto, 4200-1353 Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-1353 Porto, Portugal
| | - Augusta Monteiro
- Instituto de Investigaçaõ e Inovaçaõ em Saúde, Universidade do Porto, 4200-1353 Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-1353 Porto, Portugal
| | - Claudio E. Sunkel
- Instituto de Investigaçaõ e Inovaçaõ em Saúde, Universidade do Porto, 4200-1353 Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-1353 Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-353 Porto, Portugal
| | - Luís Pedro Resende
- Instituto de Investigaçaõ e Inovaçaõ em Saúde, Universidade do Porto, 4200-1353 Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-1353 Porto, Portugal
| |
Collapse
|
22
|
Conditional CRISPR-Cas Genome Editing in Drosophila to Generate Intestinal Tumors. Cells 2021; 10:cells10113156. [PMID: 34831379 PMCID: PMC8620722 DOI: 10.3390/cells10113156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 01/19/2023] Open
Abstract
CRISPR-Cas has revolutionized genetics and extensive efforts have been made to enhance its editing efficiency by developing increasingly more elaborate tools. Here, we evaluate the CRISPR-Cas9 system in Drosophila melanogaster to assess its ability to induce stem cell-derived tumors in the intestine. We generated conditional tissue-specific CRISPR knockouts using different Cas9 expression vectors with guide RNAs targeting the BMP, Notch, and JNK pathways in intestinal progenitors such as stem cells (ISCs) and enteroblasts (EBs). Perturbing Notch and BMP signaling increased the proliferation of ISCs/EBs and resulted in the formation of intestinal tumors, albeit with different efficiencies. By assessing both the anterior and posterior regions of the midgut, we observed regional differences in ISC/EB proliferation and tumor formation upon mutagenesis. Surprisingly, high continuous expression of Cas9 in ISCs/EBs blocked age-dependent increase in ISCs/EBs proliferation and when combined with gRNAs targeting tumor suppressors, it prevented tumorigenesis. However, no such effects were seen when temporal parameters of Cas9 were adjusted to regulate its expression levels or with a genetically modified version, which expresses Cas9 at lower levels, suggesting that fine-tuning Cas9 expression is essential to avoid deleterious effects. Our findings suggest that modifications to Cas9 expression results in differences in editing efficiency and careful considerations are required when choosing reagents for CRISPR-Cas9 mutagenesis studies. In summary, Drosophila can serve as a powerful model for context-dependent CRISPR-Cas based perturbations and to test genome-editing systems in vivo.
Collapse
|
23
|
Proske A, Bossen J, von Frieling J, Roeder T. Low-protein diet applied as part of combination therapy or stand-alone normalizes lifespan and tumor proliferation in a model of intestinal cancer. Aging (Albany NY) 2021; 13:24017-24036. [PMID: 34766923 PMCID: PMC8610115 DOI: 10.18632/aging.203692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/26/2021] [Indexed: 12/27/2022]
Abstract
Tumors of the intestinal tract are among the most common tumor diseases in humans, but, like many other tumor entities, show an unsatisfactory prognosis with a need for effective therapies. To test whether nutritional interventions and a combination with a targeted therapy can effectively cure these cancers, we used the fruit fly Drosophila as a model. In this system, we induced tumors by EGFR overexpression in intestinal stem cells. Limiting the amount of protein in the diet restored life span to that of control animals. In combination with a specific EGFR inhibitor, all major tumor-associated phenotypes could be rescued. This form of treatment was also successful in a real treatment scenario, which means when they started after the full tumor phenotype was expressed. In conclusion, reduced protein administration can be a very promising form of adjuvant cancer therapy.
Collapse
Affiliation(s)
- Alina Proske
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany
| | - Judith Bossen
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Jakob von Frieling
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany
| | - Thomas Roeder
- Department of Molecular Physiology, Zoological Institute, Kiel University, Kiel, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| |
Collapse
|
24
|
Ishibashi JR, Keshri R, Taslim TH, Brewer DK, Chan TC, Lyons S, McManamen AM, Chen A, Del Castillo D, Ruohola-Baker H. Chemical Genetic Screen in Drosophila Germline Uncovers Small Molecule Drugs That Sensitize Stem Cells to Insult-Induced Apoptosis. Cells 2021; 10:cells10102771. [PMID: 34685753 PMCID: PMC8534514 DOI: 10.3390/cells10102771] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
Cancer stem cells, in contrast to their more differentiated daughter cells, can endure genotoxic insults, escape apoptosis, and cause tumor recurrence. Understanding how normal adult stem cells survive and go to quiescence may help identify druggable pathways that cancer stem cells have co-opted. In this study, we utilize a genetically tractable model for stem cell survival in the Drosophila gonad to screen drug candidates and probe chemical-genetic interactions. Our study employs three levels of small molecule screening: (1) a medium-throughput primary screen in male germline stem cells (GSCs), (2) a secondary screen with irradiation and protein-constrained food in female GSCs, and (3) a tertiary screen in breast cancer organoids in vitro. Herein, we uncover a series of small molecule drug candidates that may sensitize cancer stem cells to apoptosis. Further, we have assessed these small molecules for chemical-genetic interactions in the germline and identified the NF-κB pathway as an essential and druggable pathway in GSC quiescence and viability. Our study demonstrates the power of the Drosophila stem cell niche as a model system for targeted drug discovery.
Collapse
Affiliation(s)
- Julien Roy Ishibashi
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Riya Keshri
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Tommy Henry Taslim
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Daniel Kennedy Brewer
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Tung Ching Chan
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Scott Lyons
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Anika Marie McManamen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Ashley Chen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Debra Del Castillo
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; (J.R.I.); (R.K.); (T.H.T.); (D.K.B.); (T.C.C.); (S.L.); (A.M.M.); (A.C.); (D.D.C.)
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
- Correspondence:
| |
Collapse
|
25
|
Ding G, Xiang X, Hu Y, Xiao G, Chen Y, Binari R, Comjean A, Li J, Rushworth E, Fu Z, Mohr SE, Perrimon N, Song W. Coordination of tumor growth and host wasting by tumor-derived Upd3. Cell Rep 2021; 36:109553. [PMID: 34407411 PMCID: PMC8410949 DOI: 10.1016/j.celrep.2021.109553] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 06/04/2021] [Accepted: 07/27/2021] [Indexed: 11/25/2022] Open
Abstract
yki-induced gut tumors in Drosophila are associated with host wasting, including muscle dysfunction, lipid loss, and hyperglycemia, a condition reminiscent of human cancer cachexia. We previously used this model to identify tumor-derived ligands that contribute to host wasting. To identify additional molecular networks involved in host-tumor interactions, we develop PathON, a web-based tool analyzing the major signaling pathways in Drosophila, and uncover the Upd3/Jak/Stat axis as an important modulator. We find that yki-gut tumors secrete Upd3 to promote self-overproliferation and enhance Jak/Stat signaling in host organs to cause wasting, including muscle dysfunction, lipid loss, and hyperglycemia. We further reveal that Upd3/Jak/Stat signaling in the host organs directly triggers the expression of ImpL2, an antagonistic binding protein for insulin-like peptides, to impair insulin signaling and energy balance. Altogether, our results demonstrate that yki-gut tumors produce a Jak/Stat pathway ligand, Upd3, that regulates both self-growth and host wasting. Ding et al. show that yki3SA-gut tumors produce Upd3 as a cachectic ligand to simultaneously promote self-growth and host organ wasting via systemic activation of Jak/Stat signaling in Drosophila. The Upd3/Jak/Stat axis induces host ImpL2 production and perturbs insulin response, leading to muscle mitochondrial dysfunction, lipid loss, and carbohydrate elevation.
Collapse
Affiliation(s)
- Guangming Ding
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei 430071, PR China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Xiaoxiang Xiang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei 430071, PR China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Gen Xiao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei 430071, PR China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Yuchen Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei 430071, PR China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Richard Binari
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Aram Comjean
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jiaying Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei 430071, PR China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Elisabeth Rushworth
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei 430071, PR China
| | - Zhenming Fu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Stephanie E Mohr
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, PR China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei 430071, PR China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
26
|
Gondal MN, Butt RN, Shah OS, Sultan MU, Mustafa G, Nasir Z, Hussain R, Khawar H, Qazi R, Tariq M, Faisal A, Chaudhary SU. A Personalized Therapeutics Approach Using an In Silico Drosophila Patient Model Reveals Optimal Chemo- and Targeted Therapy Combinations for Colorectal Cancer. Front Oncol 2021; 11:692592. [PMID: 34336681 PMCID: PMC8323493 DOI: 10.3389/fonc.2021.692592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
In silico models of biomolecular regulation in cancer, annotated with patient-specific gene expression data, can aid in the development of novel personalized cancer therapeutic strategies. Drosophila melanogaster is a well-established animal model that is increasingly being employed to evaluate such preclinical personalized cancer therapies. Here, we report five Boolean network models of biomolecular regulation in cells lining the Drosophila midgut epithelium and annotate them with colorectal cancer patient-specific mutation data to develop an in silico Drosophila Patient Model (DPM). We employed cell-type-specific RNA-seq gene expression data from the FlyGut-seq database to annotate and then validate these networks. Next, we developed three literature-based colorectal cancer case studies to evaluate cell fate outcomes from the model. Results obtained from analyses of the proposed DPM help: (i) elucidate cell fate evolution in colorectal tumorigenesis, (ii) validate cytotoxicity of nine FDA-approved CRC drugs, and (iii) devise optimal personalized treatment combinations. The personalized network models helped identify synergistic combinations of paclitaxel-regorafenib, paclitaxel-bortezomib, docetaxel-bortezomib, and paclitaxel-imatinib for treating different colorectal cancer patients. Follow-on therapeutic screening of six colorectal cancer patients from cBioPortal using this drug combination demonstrated a 100% increase in apoptosis and a 100% decrease in proliferation. In conclusion, this work outlines a novel roadmap for decoding colorectal tumorigenesis along with the development of personalized combinatorial therapeutics for preclinical translational studies.
Collapse
Affiliation(s)
- Mahnoor Naseer Gondal
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rida Nasir Butt
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Osama Shiraz Shah
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Umer Sultan
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Ghulam Mustafa
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Zainab Nasir
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Risham Hussain
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Huma Khawar
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Romena Qazi
- Department of Pathology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Muhammad Tariq
- Epigenetics Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Amir Faisal
- Cancer Therapeutics Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| | - Safee Ullah Chaudhary
- Biomedical Informatics Research Laboratory, Department of Biology, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
27
|
Sensitive High-Throughput Assays for Tumour Burden Reveal the Response of a Drosophila melanogaster Model of Colorectal Cancer to Standard Chemotherapies. Int J Mol Sci 2021; 22:ijms22105101. [PMID: 34065887 PMCID: PMC8151205 DOI: 10.3390/ijms22105101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/30/2021] [Accepted: 05/05/2021] [Indexed: 01/04/2023] Open
Abstract
Drosophila melanogaster (Drosophila) models of cancer are emerging as powerful tools to investigate the basic mechanisms underlying tumour progression and identify novel therapeutics. Rapid and inexpensive, it is possible to carry out genetic and drug screens at a far larger scale than in vertebrate organisms. Such whole-organism-based drug screens permits assessment of drug absorption and toxicity, reducing the possibility of false positives. Activating mutations in the Wnt and Ras signalling pathways are common in many epithelial cancers, and when driven in the adult Drosophila midgut, it induces aggressive intestinal tumour-like outgrowths that recapitulate many aspects of human colorectal cancer (CRC). Here we have taken a Drosophila CRC model in which tumourous cells are marked with both GFP and luciferase reporter genes, and developed novel high-throughput assays for quantifying tumour burden. Leveraging these assays, we find that the Drosophila CRC model responds rapidly to treatment with standard CRC-drugs, opening the door to future rapid genetic and drug screens.
Collapse
|
28
|
Affiliation(s)
- Helena E. Richardson
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Centre, 7 St Andrew's place, East Melbourne, Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, 7 St Andrew's place, East Melbourne, Melbourne, Victoria, 3002, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, 1-100 Grattan street, Parkville, Melbourne, Victoria, 3010, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, 1-100 Grattan street, Parkville, Melbourne, Victoria, 3010, Australia
| |
Collapse
|
29
|
Tamamouna V, Rahman MM, Petersson M, Charalambous I, Kux K, Mainor H, Bolender V, Isbilir B, Edgar BA, Pitsouli C. Remodelling of oxygen-transporting tracheoles drives intestinal regeneration and tumorigenesis in Drosophila. Nat Cell Biol 2021; 23:497-510. [PMID: 33972730 PMCID: PMC8567841 DOI: 10.1038/s41556-021-00674-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 03/31/2021] [Indexed: 02/03/2023]
Abstract
The Drosophila trachea, as the functional equivalent of mammalian blood vessels, senses hypoxia and oxygenates the body. Here, we show that the adult intestinal tracheae are dynamic and respond to enteric infection, oxidative agents and tumours with increased terminal branching. Increased tracheation is necessary for efficient damage-induced intestinal stem cell (ISC)-mediated regeneration and is sufficient to drive ISC proliferation in undamaged intestines. Gut damage or tumours induce HIF-1α (Sima in Drosophila), which stimulates tracheole branching via the FGF (Branchless (Bnl))-FGFR (Breathless (Btl)) signalling cascade. Bnl-Btl signalling is required in the intestinal epithelium and the trachea for efficient damage-induced tracheal remodelling and ISC proliferation. Chemical or Pseudomonas-generated reactive oxygen species directly affect the trachea and are necessary for branching and intestinal regeneration. Similarly, tracheole branching and the resulting increase in oxygenation are essential for intestinal tumour growth. We have identified a mechanism of tracheal-intestinal tissue communication, whereby damage and tumours induce neo-tracheogenesis in Drosophila, a process reminiscent of cancer-induced neoangiogenesis in mammals.
Collapse
Affiliation(s)
- Vasilia Tamamouna
- University of Cyprus, Department of Biological Sciences, 1 Panepistimiou Avenue, 2109 Aglantzia, Cyprus
| | - M. Mahidur Rahman
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Monika Petersson
- German Cancer Research Center (DKFZ)-Center for Molecular Biology (ZMBH), University of Heidelberg Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Irini Charalambous
- University of Cyprus, Department of Biological Sciences, 1 Panepistimiou Avenue, 2109 Aglantzia, Cyprus
| | - Kristina Kux
- University of Cyprus, Department of Biological Sciences, 1 Panepistimiou Avenue, 2109 Aglantzia, Cyprus
| | - Hannah Mainor
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Verena Bolender
- German Cancer Research Center (DKFZ)-Center for Molecular Biology (ZMBH), University of Heidelberg Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Buse Isbilir
- German Cancer Research Center (DKFZ)-Center for Molecular Biology (ZMBH), University of Heidelberg Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Bruce A. Edgar
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA,Corresponding authors ,
| | - Chrysoula Pitsouli
- University of Cyprus, Department of Biological Sciences, 1 Panepistimiou Avenue, 2109 Aglantzia, Cyprus,Corresponding authors ,
| |
Collapse
|
30
|
Parkhitko AA, Singh A, Hsieh S, Hu Y, Binari R, Lord CJ, Hannenhalli S, Ryan CJ, Perrimon N. Cross-species identification of PIP5K1-, splicing- and ubiquitin-related pathways as potential targets for RB1-deficient cells. PLoS Genet 2021; 17:e1009354. [PMID: 33591981 PMCID: PMC7909629 DOI: 10.1371/journal.pgen.1009354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/26/2021] [Accepted: 01/11/2021] [Indexed: 01/02/2023] Open
Abstract
The RB1 tumor suppressor is recurrently mutated in a variety of cancers including retinoblastomas, small cell lung cancers, triple-negative breast cancers, prostate cancers, and osteosarcomas. Finding new synthetic lethal (SL) interactions with RB1 could lead to new approaches to treating cancers with inactivated RB1. We identified 95 SL partners of RB1 based on a Drosophila screen for genetic modifiers of the eye phenotype caused by defects in the RB1 ortholog, Rbf1. We validated 38 mammalian orthologs of Rbf1 modifiers as RB1 SL partners in human cancer cell lines with defective RB1 alleles. We further show that for many of the RB1 SL genes validated in human cancer cell lines, low activity of the SL gene in human tumors, when concurrent with low levels of RB1 was associated with improved patient survival. We investigated higher order combinatorial gene interactions by creating a novel Drosophila cancer model with co-occurring Rbf1, Pten and Ras mutations, and found that targeting RB1 SL genes in this background suppressed the dramatic tumor growth and rescued fly survival whilst having minimal effects on wild-type cells. Finally, we found that drugs targeting the identified RB1 interacting genes/pathways, such as UNC3230, PYR-41, TAK-243, isoginkgetin, madrasin, and celastrol also elicit SL in human cancer cell lines. In summary, we identified several high confidence, evolutionarily conserved, novel targets for RB1-deficient cells that may be further adapted for the treatment of human cancer.
Collapse
Affiliation(s)
- Andrey A. Parkhitko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Arashdeep Singh
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sharon Hsieh
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard Binari
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Christopher J. Lord
- CRUK Gene Function Laboratory, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Colm J. Ryan
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
- School of Computer Science, University College Dublin, Dublin, Ireland
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| |
Collapse
|
31
|
Jonckheere S, Adams J, De Groote D, Campbell K, Berx G, Goossens S. Epithelial-Mesenchymal Transition (EMT) as a Therapeutic Target. Cells Tissues Organs 2021; 211:157-182. [PMID: 33401271 DOI: 10.1159/000512218] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/11/2020] [Indexed: 11/19/2022] Open
Abstract
Metastasis is the spread of cancer cells from the primary tumour to distant sites and organs throughout the body. It is the primary cause of cancer morbidity and mortality, and is estimated to account for 90% of cancer-related deaths. During the initial steps of the metastatic cascade, epithelial cancer cells undergo an epithelial-mesenchymal transition (EMT), and as a result become migratory and invasive mesenchymal-like cells while acquiring cancer stem cell properties and therapy resistance. As EMT is involved in such a broad range of processes associated with malignant transformation, it has become an increasingly interesting target for the development of novel therapeutic strategies. Anti-EMT therapeutic strategies could potentially not only prevent the invasion and dissemination of cancer cells, and as such prevent the formation of metastatic lesions, but also attenuate cancer stemness and increase the effectiveness of more classical chemotherapeutics. In this review, we give an overview about the pros and cons of therapies targeting EMT and discuss some already existing candidate drug targets and high-throughput screening tools to identify novel anti-EMT compounds.
Collapse
Affiliation(s)
- Sven Jonckheere
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jamie Adams
- Department of Biomedical Science, The University of Sheffield, Sheffield, United Kingdom
| | - Dominic De Groote
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Kyra Campbell
- Department of Biomedical Science, The University of Sheffield, Sheffield, United Kingdom
| | - Geert Berx
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium, .,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium,
| |
Collapse
|
32
|
Momtaz AZ, Ahumada Sabagh AD, Gonzalez Amortegui JG, Salazar SA, Finessi A, Hernandez J, Christensen S, Serbus LR. A Role for Maternal Factors in Suppressing Cytoplasmic Incompatibility. Front Microbiol 2020; 11:576844. [PMID: 33240234 PMCID: PMC7680759 DOI: 10.3389/fmicb.2020.576844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/20/2020] [Indexed: 12/02/2022] Open
Abstract
Wolbachia are maternally transmitted bacterial endosymbionts, carried by approximately half of all insect species. Wolbachia prevalence in nature stems from manipulation of host reproduction to favor the success of infected females. The best known reproductive modification induced by Wolbachia is referred to as sperm-egg Cytoplasmic Incompatibility (CI). In CI, the sperm of Wolbachia-infected males cause embryonic lethality, attributed to paternal chromatin segregation defects during early mitotic divisions. Remarkably, the embryos of Wolbachia-infected females “rescue” CI lethality, yielding egg hatch rates equivalent to uninfected female crosses. Several models have been discussed as the basis for Rescue, and functional evidence indicates a major contribution by Wolbachia CI factors. A role for host contributions to Rescue remains largely untested. In this study, we used a chemical feeding approach to test for CI suppression capabilities by Drosophila simulans. We found that uninfected females exhibited significantly higher CI egg hatch rates in response to seven chemical treatments that affect DNA integrity, cell cycle control, and protein turnover. Three of these treatments suppressed CI induced by endogenous wRi Wolbachia, as well as an ectopic wMel Wolbachia infection. The results implicate DNA integrity as a focal aspect of CI suppression for different Wolbachia strains. The framework presented here, applied to diverse CI models, will further enrich our understanding of host reproductive manipulation by insect endosymbionts.
Collapse
Affiliation(s)
- Ajm Zehadee Momtaz
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Abraham D Ahumada Sabagh
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Julian G Gonzalez Amortegui
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Samuel A Salazar
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Andrea Finessi
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Jethel Hernandez
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Steen Christensen
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Laura R Serbus
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| |
Collapse
|
33
|
Zhang M, Nagaosa K, Nakai Y, Yasugi T, Kushihiki M, Rahmatika D, Sato M, Shiratsuchi A, Nakanishi Y. Role for phagocytosis in the prevention of neoplastic transformation in Drosophila. Genes Cells 2020; 25:675-684. [PMID: 32865275 DOI: 10.1111/gtc.12804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 08/18/2020] [Accepted: 08/22/2020] [Indexed: 01/08/2023]
Abstract
Immunity is considered to be involved in the prevention of cancer. Although both humoral and cellular immune reactions may participate, underlying mechanisms have yet to be clarified. The present study was conducted to clarify this issue using a Drosophila model, in which neoplastic transformation was induced through the simultaneous inhibition of cell-cycle checkpoints and apoptosis. We first determined the location of hemocytes, blood cells of Drosophila playing a role of immune cells, in neoplasia-induced and normal larvae, but there was no significant difference between the two groups. When gene expression pattern in larval hemocytes was determined, the expression of immunity-related genes including those necessary for phagocytosis was reduced in the neoplasia model. We then asked the involvement of phagocytosis in the prevention of neoplasia examining animals where the expression of engulfment receptors instead of apoptosis was retarded. We found that the inhibition of engulfment receptor expression augmented the occurrence of neoplasia induced by a defect in cell-cycle checkpoints. This suggested a role for phagocytosis in the prevention of neoplastic transformation in Drosophila.
Collapse
Affiliation(s)
- Min Zhang
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kaz Nagaosa
- Section of Food Sciences, Institute of Regional Innovation, Hirosaki University, Aomori, Japan
| | - Yuji Nakai
- Section of Food Sciences, Institute of Regional Innovation, Hirosaki University, Aomori, Japan
| | - Tetsuo Yasugi
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Masako Kushihiki
- Section of Food Sciences, Institute of Regional Innovation, Hirosaki University, Aomori, Japan
| | - Dini Rahmatika
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Akiko Shiratsuchi
- Department of Liberal Arts and Sciences, Center for Medical Education, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
34
|
Bajpai A, Quazi TA, Tang HW, Manzar N, Singh V, Thakur A, Ateeq B, Perrimon N, Sinha P. A Drosophila model of oral peptide therapeutics for adult intestinal stem cell tumors. Dis Model Mech 2020; 13:dmm044420. [PMID: 32540914 PMCID: PMC7390633 DOI: 10.1242/dmm.044420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/27/2020] [Indexed: 11/20/2022] Open
Abstract
Peptide therapeutics, unlike small-molecule drugs, display crucial advantages of target specificity and the ability to block large interacting interfaces, such as those of transcription factors. The transcription co-factor of the Hippo pathway, YAP/Yorkie (Yki), has been implicated in many cancers, and is dependent on its interaction with the DNA-binding TEAD/Sd proteins via a large Ω-loop. In addition, the mammalian vestigial-like (VGLL) proteins, specifically their TONDU domain, competitively inhibit YAP-TEAD interaction, resulting in arrest of tumor growth. Here, we show that overexpression of the TONDU peptide or its oral uptake leads to suppression of Yki-driven intestinal stem cell tumors in the adult Drosophila midgut. In addition, comparative proteomic analyses of peptide-treated and untreated tumors, together with chromatin immunoprecipitation analysis, reveal that integrin pathway members are part of the Yki-oncogenic network. Collectively, our findings establish Drosophila as a reliable in vivo platform to screen for cancer oral therapeutic peptides and reveal a tumor suppressive role for integrins in Yki-driven tumors.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Anjali Bajpai
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Taushif Ahmad Quazi
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Hong-Wen Tang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nishat Manzar
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Virender Singh
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ashwani Thakur
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Bushra Ateeq
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Pradip Sinha
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
35
|
Al Outa A, Abubaker D, Madi J, Nasr R, Shirinian M. The Leukemic Fly: Promises and Challenges. Cells 2020; 9:E1737. [PMID: 32708107 PMCID: PMC7409271 DOI: 10.3390/cells9071737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 11/17/2022] Open
Abstract
Leukemia involves different types of blood cancers, which lead to significant mortality and morbidity. Murine models of leukemia have been instrumental in understanding the biology of the disease and identifying therapeutics. However, such models are time consuming and expensive in high throughput genetic and drug screening. Drosophilamelanogaster has emerged as an invaluable in vivo model for studying different diseases, including cancer. Fruit flies possess several hematopoietic processes and compartments that are in close resemblance to their mammalian counterparts. A number of studies succeeded in characterizing the fly's response upon the expression of human leukemogenic proteins in hematopoietic and non-hematopoietic tissues. Moreover, some of these studies showed that these models are amenable to genetic screening. However, none were reported to be tested for drug screening. In this review, we describe the Drosophila hematopoietic system, briefly focusing on leukemic diseases in which fruit flies have been used. We discuss myeloid and lymphoid leukemia fruit fly models and we further highlight their roles for future therapeutic screening. In conclusion, fruit fly leukemia models constitute an interesting area which could speed up the process of integrating new therapeutics when complemented with mammalian models.
Collapse
Affiliation(s)
- Amani Al Outa
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Dana Abubaker
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Joelle Madi
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Rihab Nasr
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Margret Shirinian
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| |
Collapse
|
36
|
Kamdem JP, Duarte AE, Ibrahim M, Lukong KE, Barros LM, Roeder T. Bibliometric analysis of personalized humanized mouse and Drosophila models for effective combinational therapy in cancer patients. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165880. [PMID: 32592936 DOI: 10.1016/j.bbadis.2020.165880] [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: 02/19/2020] [Revised: 05/25/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022]
Abstract
Research performed using model organisms such as mice and the fruit fly, Drosophila melanogaster has significantly enhanced our knowledge about cancer biology and the fundamental processes of cancer. This is because the major biological properties and genes associated with cancer including signaling pathways, oncogenes, tumor suppressors, and other regulators of cell growth and proliferation are evolutionary conserved. This review provides bibliometric analysis of research productivity, and performance of authors, institutions, countries, and journals associated with personalized animal cancer models, focussing on the role of Drosophila in cancer research, thus highlighting emerging trends in the field. A total of 1469 and 2672 original articles and reviews for Drosophila cancer model and patient-derived xenograft (PDX) respectively, were retrieved from the Scopus database and the most cited papers were thoroughly analyzed. Our analysis indicates a steadily increasing productivity of the animal models and especially of mouse models in cancer research. In addition to the many different systems that address almost all aspects of tumor research in humanized animal models, a trend towards using tailored screening platforms with Drosophila models in particular will become widespread in the future. Having Drosophila models that recapitulate major genetic aspects of a given tumor will enable the development and validation of novel therapeutic strategies for specific cancers, and provide a platform for screening small molecule inhibitors and other anti-tumor compounds. The combination of Drosophila cancer models and mouse PDX models particularly is highly promising and should be one of the major research strategies the future.
Collapse
Affiliation(s)
- Jean Paul Kamdem
- Department of Biological Sciences, Regional University of Cariri, Campus Pimenta, Crato, Ceara CEP: 63105-000, Brazil.
| | - Antonia Eliene Duarte
- Department of Biological Sciences, Regional University of Cariri, Campus Pimenta, Crato, Ceara CEP: 63105-000, Brazil
| | - Mohammad Ibrahim
- Department of Chemistry, Abdul Wali Khan University Mardan (AWKUM), KPK, Mardan, Pakistan
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology and Immunology (BMI) College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| | - Luiz Marivando Barros
- Department of Biological Sciences, Regional University of Cariri, Campus Pimenta, Crato, Ceara CEP: 63105-000, Brazil
| | - Thomas Roeder
- Christian-Albrechts Universität zu Kiel, Zoologisches Institut, Molekulare Physiologie, Olshausenstraße 40, D-24098 Kiel, Germany; German Center for Lung Research, Airway Research Center North, Kiel, Germany.
| |
Collapse
|
37
|
Liang Q, Ma P, Zhang Q, Yin Y, Wang P, Wang S, Zhang Y, Han R, Deng H. A gum Arabic assisted sustainable drug delivery system for adult Drosophila. Biol Open 2020; 9:bio052241. [PMID: 32487516 PMCID: PMC7328006 DOI: 10.1242/bio.052241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/11/2020] [Indexed: 01/26/2023] Open
Abstract
Large-scale compound screening in adult flies is hampered by the lack of continuous drug delivery systems and poor solubility of numerous compounds. Here we found that gum Arabic (Acacia/Senegal gum), a widely used stabilizer, can also emulsify lipophilic compounds and profoundly increase their accessibility to target tissues in Drosophila and mice. We further developed a gum Arabic-based drug delivery system, wherein the drug was ground into gum Arabic and emulsified in liquid food fed to flies by siphoning through a U-shape glass capillary. This system did not affect food intake nor cell viability. Since drugs were continuously delivered by siphoning, minimal compound waste and less frequent food changes make this system ideal for large-scale long-term screenings. In our pilot screening for antitumor drugs in the NCI DTP library, we used a Drosophila model of colorectal cancer and identified two drugs that are especially hydrophobic and were not identified in previous screenings. Our data demonstrated that gum Arabic facilitates drug delivery in animal models and the system is suitable for long-term high-throughput drug screening in Drosophila This system would accelerate drug discovery for chronic and cognitive conditions.
Collapse
Affiliation(s)
- Qiying Liang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
- College of Animal Sciences and Technology, Guangxi University, Nanning, 530004, China
| | - Peng Ma
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| | - Qi Zhang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| | - Youjie Yin
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| | - Ping Wang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| | - Saifei Wang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| | - Yao Zhang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| | - Ruolei Han
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| | - Hansong Deng
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Building, 1239 Siping Road, Yangpu District, 20092 China
| |
Collapse
|
38
|
von Frieling J, Faisal MN, Sporn F, Pfefferkorn R, Nolte SS, Sommer F, Rosenstiel P, Roeder T. A high-fat diet induces a microbiota-dependent increase in stem cell activity in the Drosophila intestine. PLoS Genet 2020; 16:e1008789. [PMID: 32453733 PMCID: PMC7274450 DOI: 10.1371/journal.pgen.1008789] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/05/2020] [Accepted: 04/22/2020] [Indexed: 12/25/2022] Open
Abstract
Over-consumption of high-fat diets (HFDs) is associated with several pathologies. Although the intestine is the organ that comes into direct contact with all diet components, the impact of HFD has mostly been studied in organs that are linked to obesity and obesity related disorders. We used Drosophila as a simple model to disentangle the effects of a HFD on the intestinal structure and physiology from the plethora of other effects caused by this nutritional intervention. Here, we show that a HFD, composed of triglycerides with saturated fatty acids, triggers activation of intestinal stem cells in the Drosophila midgut. This stem cell activation was transient and dependent on the presence of an intestinal microbiota, as it was completely absent in germ free animals. Moreover, major components of the signal transduction pathway have been elucidated. Here, JNK (basket) in enterocytes was necessary to trigger synthesis of the cytokine upd3 in these cells. This ligand in turn activated the JAK/STAT pathway in intestinal stem cells. Chronic subjection to a HFD markedly altered both the microbiota composition and the bacterial load. Although HFD-induced stem cell activity was transient, long-lasting changes to the cellular composition, including a substantial increase in the number of enteroendocrine cells, were observed. Taken together, a HFD enhances stem cell activity in the Drosophila gut and this effect is completely reliant on the indigenous microbiota and also dependent on JNK signaling within intestinal enterocytes. High-fat diets have been associated with a plethora of morbidities. The major research focus has been on its effects on obesity related disorders, mostly omitting the intestine, although it is the organ that makes the first contact with all diet components. Here, we aimed to understand the effects of HFD on the intestine itself. Using Drosophila as a model, we showed that a HFD and more specifically, trigylcerides with saturated fatty acids, induced a transient activation of intestinal stem cells. This response completely depended on the presence of an intestinal microbiota, as in germ free flies this reaction was completely abolished. Mechanistically, we found that HFD induces JNK signaling in enterocytes, which triggers production of the cytokine upd3. This ligand of the JAK/STAT pathway, in turn activates STAT signaling in intestinal stem cells, leading to their activation. All these components of the JNK- and JAK/STAT-pathways are necessary for a HFD to lead to increased stem cell production. Moreover, HFD changed both, composition and abundance of the microbiota. As fecal transfer experiments failed to recapitulate the HFD phenotype, we assume that the increased bacterial load is the major cause for the HFD triggered stem cell activation in the intestine.
Collapse
Affiliation(s)
- Jakob von Frieling
- Zoological Institute, Department of Molecular Physiology, Kiel University, Kiel, Germany
| | - Muhammed Naeem Faisal
- Zoological Institute, Department of Molecular Physiology, Kiel University, Kiel, Germany
| | - Femke Sporn
- Zoological Institute, Department of Molecular Physiology, Kiel University, Kiel, Germany
| | - Roxana Pfefferkorn
- Zoological Institute, Department of Molecular Physiology, Kiel University, Kiel, Germany
| | - Stella Solveig Nolte
- Zoological Institute, Department of Molecular Physiology, Kiel University, Kiel, Germany
| | | | | | - Thomas Roeder
- Zoological Institute, Department of Molecular Physiology, Kiel University, Kiel, Germany
- German Center for Lung Research, Airway Research Center North, Kiel, Germany
- * E-mail:
| |
Collapse
|
39
|
Strategies for Functional Interrogation of Big Cancer Data Using Drosophila Cancer Models. Int J Mol Sci 2020; 21:ijms21113754. [PMID: 32466549 PMCID: PMC7312059 DOI: 10.3390/ijms21113754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Rapid development of high throughput genome analysis technologies accompanied by significant reduction in costs has led to the accumulation of an incredible amount of data during the last decade. The emergence of big data has had a particularly significant impact in biomedical research by providing unprecedented, systems-level access to many disease states including cancer, and has created promising opportunities as well as new challenges. Arguably, the most significant challenge cancer research currently faces is finding effective ways to use big data to improve our understanding of molecular mechanisms underlying tumorigenesis and developing effective new therapies. Functional exploration of these datasets and testing predictions from computational approaches using experimental models to interrogate their biological relevance is a key step towards achieving this goal. Given the daunting scale and complexity of the big data available, experimental systems like Drosophila that allow large-scale functional studies and complex genetic manipulations in a rapid, cost-effective manner will be of particular importance for this purpose. Findings from these large-scale exploratory functional studies can then be used to formulate more specific hypotheses to be explored in mammalian models. Here, I will discuss several strategies for functional exploration of big cancer data using Drosophila cancer models.
Collapse
|
40
|
Drosophila as a model for studying cystic fibrosis pathophysiology of the gastrointestinal system. Proc Natl Acad Sci U S A 2020; 117:10357-10367. [PMID: 32345720 DOI: 10.1073/pnas.1913127117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF) is a recessive disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The most common symptoms include progressive lung disease and chronic digestive conditions. CF is the first human genetic disease to benefit from having five different species of animal models. Despite the phenotypic differences among the animal models and human CF, these models have provided invaluable insight into understanding disease mechanisms at the organ-system level. Here, we identify a member of the ABCC4 family, CG5789, that has the structural and functional properties expected for encoding the Drosophila equivalent of human CFTR, and thus refer to it as Drosophila CFTR (Dmel\CFTR). We show that knockdown of Dmel\CFTR in the adult intestine disrupts osmotic homeostasis and displays CF-like phenotypes that lead to intestinal stem cell hyperplasia. We also show that expression of wild-type human CFTR, but not mutant variants of CFTR that prevent plasma membrane expression, rescues the mutant phenotypes of Dmel\CFTR Furthermore, we performed RNA sequencing (RNA-Seq)-based transcriptomic analysis using Dmel\CFTR fly intestine and identified a mucin gene, Muc68D, which is required for proper intestinal barrier protection. Altogether, our findings suggest that Drosophila can be a powerful model organism for studying CF pathophysiology.
Collapse
|
41
|
Mahapatra K, Ghosh AK, De S, Ghosh N, Sadhukhan P, Chatterjee S, Ghosh R, Sil PC, Roy S. Assessment of cytotoxic and genotoxic potentials of a mononuclear Fe(II) Schiff base complex with photocatalytic activity in Trigonella. Biochim Biophys Acta Gen Subj 2020; 1864:129503. [PMID: 31816347 DOI: 10.1016/j.bbagen.2019.129503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND In recent times, coordination complexes of iron in various oxidation states along with variety of ligand systems have been designed and developed for effective treatment of cancer cells without adversely affecting the normal cell and tissues of various organs. METHODS In this study, we have evaluated the mechanism of action of a Fe(II) Schiff base complex in the crop plant Trigonella foenum-graecum L. (Fenugreek) as the screening system by using morphological, cytological, biochemical and molecular approaches. Further functional characterization was performed using MCF-7 cell line and solid tumour model for the assessment of anti-tumour activity of the complex. RESULTS Our results indicate efficiency of the Fe(II) Schiff base complex in the induction of double strand breaks in DNA. Complex treatment clearly induced cytotoxic and genotoxic damage in Trigonella seedlings. The Fe-complex treatment caused cell cycle arrest via the activation of ATM-ATR kinase mediated DNA damage response pathway with the compromised expression of CDK1, CDK2 and CyclinB1 protein in Trigonella seedlings. In cultured MCF-7 cells, the complex induces cytotoxicity and DNA fragmentation through intracellular ROS generation. Fe-complex treatment inhibited tumour growth in solid tumour model with no additional side effects. CONCLUSION The growth inhibitory and cytotoxic effects of the complex result from activation of DNA damage response along with oxidative stress and cell cycle arrest. GENERAL SIGNIFICANCE Overall, our results have provided comprehensive information on the mechanism of action and efficacy of a Fe(II) Schiff base complex in higher eukaryotic genomes and indicated its future implications as potential therapeutic agent.
Collapse
Affiliation(s)
- Kalyan Mahapatra
- Department of Botany, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Ayon Kanti Ghosh
- Department of Chemistry, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Sayanti De
- Department of Botany, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Noyel Ghosh
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Pritam Sadhukhan
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Sharmistha Chatterjee
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Rajarshi Ghosh
- Department of Chemistry, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Centenary Campus, P-1/12 C.I.T. Scheme VIIM, Kolkata 700054, India
| | - Sujit Roy
- Department of Botany, UGC Centre for Advanced Studies, The University of Burdwan, Golapbag Campus, Burdwan 713104, West Bengal, India.
| |
Collapse
|
42
|
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: 1.0] [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
|
43
|
Fereres S, Hatori R, Hatori M, Kornberg TB. Cytoneme-mediated signaling essential for tumorigenesis. PLoS Genet 2019; 15:e1008415. [PMID: 31568500 PMCID: PMC6786653 DOI: 10.1371/journal.pgen.1008415] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/10/2019] [Accepted: 09/11/2019] [Indexed: 11/18/2022] Open
Abstract
Communication between neoplastic cells and cells of their microenvironment is critical to cancer progression. To investigate the role of cytoneme-mediated signaling as a mechanism for distributing growth factor signaling proteins between tumor and tumor-associated cells, we analyzed EGFR and RET Drosophila tumor models and tested several genetic loss-of-function conditions that impair cytoneme-mediated signaling. Neuroglian, capricious, Irk2, SCAR, and diaphanous are genes that cytonemes require during normal development. Neuroglian and Capricious are cell adhesion proteins, Irk2 is a potassium channel, and SCAR and Diaphanous are actin-binding proteins, and the only process to which they are known to contribute jointly is cytoneme-mediated signaling. We observed that diminished function of any one of these genes suppressed tumor growth and increased organism survival. We also noted that EGFR-expressing tumor discs have abnormally extensive tracheation (respiratory tubes) and ectopically express Branchless (Bnl, a FGF) and FGFR. Bnl is a known inducer of tracheation that signals by a cytoneme-mediated process in other contexts, and we determined that exogenous over-expression of dominant negative FGFR suppressed tumor growth. Our results are consistent with the idea that cytonemes move signaling proteins between tumor and stromal cells and that cytoneme-mediated signaling is required for tumor growth and malignancy. The growth of many types of tumors depend on productive interactions with stromal, non-tumor neighbors, and although there is evidence that tumor and stromal cells exchange signaling proteins and growth factors that they produce, the mechanism by which these proteins move between the signaling cells has not been investigated and is not known. Our previous work has shown that normal cells make transient chemical synapses at sites where specialized filopodia called cytonemes contact signaling partners, and in this work we explore the possibility that tumors use the same mechanism to communicate with stromal cells. We show that cytoneme-mediated signaling is essential for growth of Drosophila tumors that model human EGFR over-expression and RET-driven disease. Remarkably, inhibition of cytonemes cures flies of lethal tumors.
Collapse
Affiliation(s)
- Sol Fereres
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, United States of America
| | - Ryo Hatori
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, United States of America
| | - Makiko Hatori
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, United States of America
| | - Thomas B. Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Pfeifle I, Bohnekamp J, Volkhardt A, Kirsten H, Rohwedder A, Thum A, Magin TM, Kunz M. MEK inhibitor cobimetinib rescues a dRaf mutant lethal phenotype in Drosophila melanogaster. Exp Dermatol 2019; 28:1079-1082. [PMID: 31338879 DOI: 10.1111/exd.14010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 05/31/2019] [Accepted: 07/17/2019] [Indexed: 12/01/2022]
Abstract
Since Drosophila melanogaster has proven to be a useful model system to study phenotypes of oncogenic mutations and to identify new anti-cancer drugs, we generated human BRAFV600E homologous dRaf mutant (dRafA572E ) Drosophila melanogaster strains to use these for characterisation of mutant phenotypes and exploit these phenotypes for drug testing. For mutant gene expression, the GAL4/UAS expression system was used. dRafA572E was expressed tissue-specific in the eye, epidermis, heart, wings, secretory glands and in the whole animal. Expression of dRaf A572E under the control of an eye-specific driver led to semi-lethality and a rough eye phenotype. The vast majority of other tissue-specific and ubiquitous drivers led to a lethal phenotype only. The rough eye phenotype was used to test BRAF inhibitor vemurafenib and MEK1/2 inhibitor cobimetinib. There was no phenotype rescue by this treatment. However, a significant rescue of the lethal phenotype was observed under a gut-specific driver. Here, MEK1/2 inhibitor cobimetinib rescued Drosophila larvae to reach pupal stage in 37% of cases as compared to 1% in control experiments. Taken together, the BRAFV600E homolog dRaf A572E exerts mostly lethal effects in Drosophila. Gut-specific dRaf A572E expression might in future be developed further for drug testing.
Collapse
Affiliation(s)
- Isabelle Pfeifle
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | - Jens Bohnekamp
- Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, Leipzig, Germany.,Division of Genetics, Institute of Biology, University of Leipzig, Leipzig, Germany
| | - Anna Volkhardt
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | - Holger Kirsten
- Interdisciplinary Center for Bioinformatics and Bioinformatics Group, Department of Computer Science, University of Leipzig, Leipzig, Germany
| | - Astrid Rohwedder
- Division of Genetics, Institute of Biology, University of Leipzig, Leipzig, Germany
| | - Andreas Thum
- Division of Genetics, Institute of Biology, University of Leipzig, Leipzig, Germany
| | - Thomas M Magin
- Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, Leipzig, Germany
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
45
|
Ekowati H, Arai J, Damana Putri AS, Nainu F, Shiratsuchi A, Nakanishi Y. Protective effects of Phaseolus vulgaris lectin against viral infection in Drosophila. Drug Discov Ther 2019; 11:329-335. [PMID: 29332891 DOI: 10.5582/ddt.2017.01071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Phytohemagglutinin (PHA) isolated from the family of Phaseolus vulgaris beans is a promising agent against viral infection; however, it has not yet been demonstrated in vivo. We herein investigated this issue using Drosophila as a host. Adult flies were fed lectin approximately 12 h before they were subjected to a systemic viral infection. After a fatal infection with Drosophila C virus, death was delayed and survival was longer in flies fed PHA-P, a mixture of L4, L3E1, and L2E2, than in control unfed flies. We then examined PHA-L4, anticipating subunit L as the active form, and confirmed the protective effects of this lectin at markedly lower concentrations than PHA-P. In both experiments, lectin feeding reduced the viral load prior to the onset of fly death. Furthermore, we found a dramatic increase in the levels of the mRNAs of phagocytosis receptors in flies after feeding with PHA-L4 while a change in the levels of the mRNAs of antimicrobial peptides was marginal. We concluded that P. vulgaris PHA protects Drosophila against viral infection by augmenting the level of host immunity.
Collapse
Affiliation(s)
- Heny Ekowati
- Graduate School of Medical Sciences, Kanazawa University.,Faculty of Health Science, Jenderal Soedirman University
| | - Junko Arai
- Product Development Laboratory, J-Oil Mills, Inc
| | | | | | | | | |
Collapse
|
46
|
Bossen J, Uliczka K, Steen L, Pfefferkorn R, Mai MMQ, Burkhardt L, Spohn M, Bruchhaus I, Fink C, Heine H, Roeder T. An EGFR-Induced Drosophila Lung Tumor Model Identifies Alternative Combination Treatments. Mol Cancer Ther 2019; 18:1659-1668. [PMID: 31217165 DOI: 10.1158/1535-7163.mct-19-0168] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/06/2019] [Accepted: 06/14/2019] [Indexed: 11/16/2022]
Abstract
Lung cancer is the leading cause of cancer-associated mortality. Mutations in the EGFR gene are among the most important inducers of lung tumor development, but success of personalized therapies is still limited because of toxicity or developing resistances. We expressed constitutively active EGFR (EGFRCA) exclusively in the airway system of Drosophila melanogaster and performed comprehensive phenotyping. Ectopic expression of EGFRCA induced massive hyper- and metaplasia, leading to early death. We used the lethal phenotype as a readout and screened a library of FDA-approved compounds and found that among the 1,000 compounds, only the tyrosine kinase inhibitors (TKI) afatinib, gefitinib, and ibrutinib rescued lethality in a whole-animal screening approach. Furthermore, we screened the library in the presence of a subtherapeutic afatinib dose and identified bazedoxifene as a synergistically acting compound that rescues EGFR-induced lethality. Our findings highlight the potential of Drosophila-based whole-animal screening approaches not only to identify specific EGFR inhibitors but also to discover compounds that act synergistically with known TKIs. Moreover, we showed that targeting the EGFR together with STAT-signaling is a promising strategy for lung tumor treatment.
Collapse
Affiliation(s)
- Judith Bossen
- Departments of Molecular Physiology and Zoology, Kiel University, Kiel, Germany
| | - Karin Uliczka
- Research Center Borstel-Leibniz Lung Center, Priority Area Asthma and Allergy, Division of Invertebrate Models, Borstel Germany.,Research Center Borstel-Leibniz Lung Center, Priority Area Asthma and Allergy, Division of Innate Immunity, Borstel, Germany
| | - Line Steen
- Departments of Molecular Physiology and Zoology, Kiel University, Kiel, Germany
| | - Roxana Pfefferkorn
- Departments of Molecular Physiology and Zoology, Kiel University, Kiel, Germany
| | - Mandy Mong-Quyen Mai
- Research Center Borstel-Leibniz Lung Center, Priority Area Asthma and Allergy, Division of Innate Immunity, Borstel, Germany
| | - Lia Burkhardt
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Next Generation Sequencing Technology Platform, Hamburg, Germany
| | - Michael Spohn
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Next Generation Sequencing Technology Platform, Hamburg, Germany
| | - Iris Bruchhaus
- Bernhard-Nocht Institute for Tropical Medicine, Dept. Parasitology, Hamburg, Germany
| | - Christine Fink
- Departments of Molecular Physiology and Zoology, Kiel University, Kiel, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Holger Heine
- Research Center Borstel-Leibniz Lung Center, Priority Area Asthma and Allergy, Division of Innate Immunity, Borstel, Germany. .,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Thomas Roeder
- Departments of Molecular Physiology and Zoology, Kiel University, Kiel, Germany. .,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf, Germany
| |
Collapse
|
47
|
Villegas SN, Gombos R, García-López L, Gutiérrez-Pérez I, García-Castillo J, Vallejo DM, Da Ros VG, Ballesta-Illán E, Mihály J, Dominguez M. PI3K/Akt Cooperates with Oncogenic Notch by Inducing Nitric Oxide-Dependent Inflammation. Cell Rep 2019. [PMID: 29514083 DOI: 10.1016/j.celrep.2018.02.049] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The PI3K/Akt signaling pathway, Notch, and other oncogenes cooperate in the induction of aggressive cancers. Elucidating how the PI3K/Akt pathway facilitates tumorigenesis by other oncogenes may offer opportunities to develop drugs with fewer side effects than those currently available. Here, using an unbiased in vivo chemical genetic screen in Drosophila, we identified compounds that inhibit the activity of proinflammatory enzymes nitric oxide synthase (NOS) and lipoxygenase (LOX) as selective suppressors of Notch-PI3K/Akt cooperative oncogenesis. Tumor silencing of NOS and LOX signaling mirrored the antitumor effect of the hit compounds, demonstrating their participation in Notch-PI3K/Akt-induced tumorigenesis. Oncogenic PI3K/Akt signaling triggered inflammation and immunosuppression via aberrant NOS expression. Accordingly, activated Notch tumorigenesis was fueled by hampering the immune response or by NOS overexpression to mimic a protumorigenic environment. Our lead compound, the LOX inhibitor BW B70C, also selectively killed human leukemic cells by dampening the NOTCH1-PI3K/AKT-eNOS axis.
Collapse
Affiliation(s)
- Santiago Nahuel Villegas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain.
| | - Rita Gombos
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Lucia García-López
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Irene Gutiérrez-Pérez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Jesús García-Castillo
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Diana Marcela Vallejo
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Vanina Gabriela Da Ros
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Esther Ballesta-Illán
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Maria Dominguez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Avda. Ramón y Cajal s/n, 03550 Sant Joan d'Alacant, Alicante, Spain.
| |
Collapse
|
48
|
Rosen H, Sharf R, Pechkovsky A, Salzberg A, Kleinberger T. Selective elimination of cancer cells by the adenovirus E4orf4 protein in a Drosophila cancer model: a new paradigm for cancer therapy. Cell Death Dis 2019; 10:455. [PMID: 31186403 PMCID: PMC6560070 DOI: 10.1038/s41419-019-1680-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 05/08/2019] [Accepted: 05/16/2019] [Indexed: 01/16/2023]
Abstract
The adenovirus (Ad) E4orf4 protein contributes to efficient progression of virus infection. When expressed alone E4orf4 induces p53- and caspase-independent cell-death, which is more effective in cancer cells than in normal cells in tissue culture. Cancer selectivity of E4orf4-induced cell-death may result from interference with various regulatory pathways that cancer cells are more dependent on, including DNA damage signaling and proliferation control. E4orf4 signaling is conserved in several organisms, including yeast, Drosophila, and mammalian cells, indicating that E4orf4-induced cell-death can be investigated in these model organisms. The Drosophila genetic model system has contributed significantly to the study of cancer and to identification of novel cancer therapeutics. Here, we used the fly model to investigate the ability of E4orf4 to eliminate cancer tissues in a whole organism with minimal damage to normal tissues. We show that E4orf4 dramatically inhibited tumorigenesis and rescued survival of flies carrying a variety of tumors, including highly aggressive and metastatic tumors in the fly brain and eye discs. Moreover, E4orf4 rescued the morphology of adult eyes containing scrib- cancer clones even when expressed at a much later stage than scrib elimination. The E4orf4 partner protein phosphatase 2A (PP2A) was required for inhibition of tumorigenesis by E4orf4 in the system described here, whereas another E4orf4 partner, Src kinase, provided only minimal contribution to this process. Our results suggest that E4orf4 is an effective anticancer agent and reveal a promising potential for E4orf4-based cancer treatments.
Collapse
Affiliation(s)
- Helit Rosen
- Department of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, 3109601, Haifa, Israel
| | - Rakefet Sharf
- Department of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, 3109601, Haifa, Israel
| | - Antonina Pechkovsky
- Department of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, 3109601, Haifa, Israel.,Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, 3109601, Haifa, Israel
| | - Adi Salzberg
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, 3109601, Haifa, Israel
| | - Tamar Kleinberger
- Department of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, 3109601, Haifa, Israel.
| |
Collapse
|
49
|
Cagan RL, Zon LI, White RM. Modeling Cancer with Flies and Fish. Dev Cell 2019; 49:317-324. [PMID: 31063751 PMCID: PMC6506185 DOI: 10.1016/j.devcel.2019.04.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/07/2019] [Accepted: 04/08/2019] [Indexed: 12/11/2022]
Abstract
Cancer has joined heart disease as the leading source of mortality in the US. In an era of organoids, patient-derived xenografts, and organs on a chip, model organisms continue to thrive with a combination of powerful genetic tools, rapid pace of discovery, and affordability. Model organisms enable the analysis of both the tumor and its associated microenvironment, aspects that are particularly relevant to our understanding of metastasis and drug resistance. In this Perspective, we explore some of the strengths of fruit flies and zebrafish for addressing fundamental cancer questions and how these two organisms can contribute to identifying promising therapeutic candidates.
Collapse
Affiliation(s)
- Ross L Cagan
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Leonard I Zon
- Children's Hospital Boston, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
50
|
Su TT. Drug screening in Drosophila; why, when, and when not? WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e346. [PMID: 31056843 DOI: 10.1002/wdev.346] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/08/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022]
Abstract
The best global seller among oncology drugs in 2018 is lenalidomide, an analog of thalidomide. It took 53 years and a circuitous route from the discovery of thalidomide to approval of an analog for use in treatment of cancer. We understand now a lot more about the genetic and molecular basis of diseases than we did in 1953 when thalidomide was discovered. We have also no shortage of chemical libraries with hundreds of thousands of compounds, both synthetic and natural. What we need are better ways to search among these rich resources for compounds with the potential to do what we want them to do. This review summarizes examples from the literature that make Drosophila melanogaster a good model to screen for drugs, and discusses knowledge gaps and technical challenges that make Drosophila models not as widely used as they could or should be. This article is categorized under: Technologies > Analysis of Cell, Tissue, and Animal Phenotypes.
Collapse
Affiliation(s)
- Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado.,Molecular, Cellular and Developmental Biology, University of Colorado Comprehensive Cancer Center, Aurora, Colorado
| |
Collapse
|