1
|
Choudhary D, Nasiruddin Khan MD, Khan Z, Mehan S, Gupta GD, Narula AS, Samant R. Navigating the complexities of neuronal signaling and targets in neurological disorders: From pathology to therapeutics. Eur J Pharmacol 2025; 995:177417. [PMID: 40010482 DOI: 10.1016/j.ejphar.2025.177417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
Neurological disorders arising from structural and functional disruptions in the nervous system present major global health challenges. This review examines the intricacies of various cellular signaling pathways, including Nrf2/Keap1/HO-1, SIRT-1, JAK/STAT3/mTOR, and BACE-1/gamma-secretase/MAPT, which play pivotal roles in neuronal health and pathology. The Nrf2-Keap1 pathway, a key antioxidant response mechanism, mitigates oxidative stress, while SIRT-1 contributes to mitochondrial integrity and inflammation control. Dysregulation of these pathways has been identified in neurodegenerative and neuropsychiatric disorders, including Alzheimer's and Parkinson's diseases, characterized by inflammation, protein aggregation, and mitochondrial dysfunction. Additionally, the JAK/STAT3 signaling pathway emphasizes the connection between cytokine responses and neuroinflammation, further compounding disease progression. This review explores the crosstalk among these signaling networks, elucidating how their disruption leads to neuronal decline. It also addresses the dual roles of these pathways, presenting challenges in targeting them for therapeutic purposes. Despite the potential benefits of activating neuroprotective pathways, excessive stimulation may cause deleterious effects, including tumorigenesis. Future research should focus on designing multi-targeted therapies that enhance the effectiveness and safety of treatments, considering individual variabilities and the obstacles posed by the blood-brain barrier to drug delivery. Understanding these complex signaling interactions is crucial for developing innovative and effective neuroprotective strategies that could significantly improve the management of neurological disorders.
Collapse
Affiliation(s)
- Divya Choudhary
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - M D Nasiruddin Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| | | |
Collapse
|
2
|
Mathews J, Erickson P, Kuchling F, Jawanda N, Pio-Lopez L, Pai VP, Levin M. Meeting Review: "National Cancer Institute Conference on Cancer Bioelectricity" September 12, 2024. Bioelectricity 2025; 7:94-104. [PMID: 40342939 PMCID: PMC12054611 DOI: 10.1089/bioe.2024.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Abstract
The Office of Cancer Complementary and Alternative Medicine, part of the Division of Cancer Treatment and Diagnosis under the National Cancer Institute (NCI), supports research in diverse areas of cancer therapeutics such as microbial therapies, herbal remedies, and mind-body practices. Recently they have become especially interested in the emerging role of bioelectricity in cancer biology and organized a virtual meeting with some of the top scientists in the field. In this report, we overview this first-of-its-kind Naional Institute of Health (NIH)-sponsored meeting, which featured talks from 14 researchers exploring the role of bioelectricity in cancer biology. The talks covered a wide range of topics, including excellent background information on how cell collectives change their bioelectrical coupling and set points during cancer formation, new tools for reading and writing bioelectrical signatures in cells and whole organisms, how ion channels that are involved in setting those signatures affect canonical pathways in development and tumor growth, and the methods for modeling bioelectrical interactions and information transfer in cell collectives. Especially exciting were the translational technologies that were highlighted, including new diagnostics, metastasis inhibition therapies, and more efficient detection of surgical margins. The meeting concluded with funding opportunities available from the NCI Division of Cancer Biology, Innovative Molecular Analysis Technologies Program, and the Small Business Innovation Research Development Center.
Collapse
Affiliation(s)
- Juanita Mathews
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| | - Patrick Erickson
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| | - Franz Kuchling
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| | - Navneet Jawanda
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| | - Léo Pio-Lopez
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| | - Vaibhav P. Pai
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
3
|
Sinnott-Armstrong N, Fields S, Roth F, Starita LM, Trapnell C, Villen J, Fowler DM, Queitsch C. Understanding genetic variants in context. eLife 2024; 13:e88231. [PMID: 39625477 PMCID: PMC11614383 DOI: 10.7554/elife.88231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 11/15/2024] [Indexed: 12/06/2024] Open
Abstract
Over the last three decades, human genetics has gone from dissecting high-penetrance Mendelian diseases to discovering the vast and complex genetic etiology of common human diseases. In tackling this complexity, scientists have discovered the importance of numerous genetic processes - most notably functional regulatory elements - in the development and progression of these diseases. Simultaneously, scientists have increasingly used multiplex assays of variant effect to systematically phenotype the cellular consequences of millions of genetic variants. In this article, we argue that the context of genetic variants - at all scales, from other genetic variants and gene regulation to cell biology to organismal environment - are critical components of how we can employ genomics to interpret these variants, and ultimately treat these diseases. We describe approaches to extend existing experimental assays and computational approaches to examine and quantify the importance of this context, including through causal analytic approaches. Having a unified understanding of the molecular, physiological, and environmental processes governing the interpretation of genetic variants is sorely needed for the field, and this perspective argues for feasible approaches by which the combined interpretation of cellular, animal, and epidemiological data can yield that knowledge.
Collapse
Affiliation(s)
- Nasa Sinnott-Armstrong
- Herbold Computational Biology Program, Fred Hutchinson Cancer CenterSeattleUnited States
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Brotman Baty Institute for Precision MedicineSeattleUnited States
| | - Stanley Fields
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Department of Medicine, University of WashingtonSeattleUnited States
| | - Frederick Roth
- Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of TorontoTorontoCanada
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai HospitalTorontoCanada
- Department of Computational and Systems Biology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Lea M Starita
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Brotman Baty Institute for Precision MedicineSeattleUnited States
| | - Cole Trapnell
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Brotman Baty Institute for Precision MedicineSeattleUnited States
| | - Judit Villen
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Brotman Baty Institute for Precision MedicineSeattleUnited States
| | - Douglas M Fowler
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Brotman Baty Institute for Precision MedicineSeattleUnited States
- Department of Bioengineering, University of WashingtonSeattleUnited States
| | - Christine Queitsch
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Brotman Baty Institute for Precision MedicineSeattleUnited States
| |
Collapse
|
4
|
Ju Y, Du M, Wang Z, Mu X, Miao Y, Guo Z, Wang D, Wang S, Ding J, Jin G, Zhang W, Qiao H, Su Y, Liu X, Yuchi Z, Tan X, Wang Y. Kukoamine A alleviates nephrolithiasis by inhibiting endogenous oxalate synthesis via the IL-6/JAK/STAT3/DAO signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156145. [PMID: 39461201 DOI: 10.1016/j.phymed.2024.156145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/16/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND The recurrent nature and socioeconomic burden of nephrolithiasis demand effective treatments. Delineating the crosstalk between inflammatory processes and the endogenous oxalate metabolism pathway, which underpins nephrolithiasis pathogenesis, is essential for advancing treatment strategies. PURPOSE We aim to screen therapeutic Chinese herbal remedies and their bioactive constituents for kidney stone treatment using a fruit fly model, followed by efficacy and mechanism validation in a rodent nephrolithiasis model as well as in vitro human cell culture model. STUDY DESIGN AND METHODS We developed a fruit fly model to screen for efficient traditional Chinese medicine herbs and their active compounds for kidney stone treatment. Candidate active compounds from efficient herbs were separated and identified by solid-phase chromatography coupled with LC-MS analysis. Fruit fly genetic tools were employed to manipulate the expression of related genes to explore the therapeutic mechanisms of the Lycii Cortex and kukoamine A (KuA). To confirm the therapeutic effects and mechanisms of KuA for mammalian nephrolithiasis, mouse model of glyoxylate-induced kidney stone and human renal tubular cells were used. The therapeutic role of kukoamine A in nephrolithiasis was evaluated by assessing tubular injury, crystal deposition, and adhesion. The level of expression and phosphorylation in cells and mice was assessed using RT-qPCR and western blot. RESULTS Our findings indicate that Lycii Cortex potently inhibits calcium oxalate stone formation via activation of the JNK/Upd3/JAK/STAT signaling cascade, resulting in diminished endogenous oxalate synthesis by downregulating D-amino acid oxidase (DAO) gene expression, predominantly in fruit fly Malpighian tube stellate cells. KuA was identified as the principal bioactive constituent mediating these effects. Both mouse models and human cell assays confirmed KuA's efficacy in preventing calcium oxalate nephrolithiasis in mammals, through hepatic JAK/STAT3 pathway activation and upregulation of IL-6, culminating in reduced urinary crystal deposition. CONCLUSION Our research underscores the potential of kukoamine A as a lead compound in treating nephrolithiasis and reveals the interplay between the IL-6/JAK/STAT3 inflammatory pathway and endogenous oxalate metabolism in nephrolithiasis pathogenesis. Additionally, it highlights the utility of the fruit fly model as a powerful tool for deciphering the therapeutic mechanisms of traditional Chinese herbs.
Collapse
Affiliation(s)
- Yingjie Ju
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Mengwei Du
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhongyi Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xin Mu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300250, China
| | - Zhimou Guo
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Dekun Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shiyao Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Junjie Ding
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Gaowa Jin
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Wen Zhang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Huanhuan Qiao
- Medical School, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Yanfang Su
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xiuyun Liu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| | - Zhiguang Yuchi
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| | - Xiaoyue Tan
- School of Medicine, Nankai University, Tianjin 300071, China.
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
5
|
Chasse AY, Bandyadka S, Wertheimer MC, Serizier SB, McCall K. Professional phagocytes are recruited for the clearance of obsolete nonprofessional phagocytes in the Drosophila ovary. Front Immunol 2024; 15:1389674. [PMID: 38994369 PMCID: PMC11236694 DOI: 10.3389/fimmu.2024.1389674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
Cell death is an important process in the body, as it occurs throughout every tissue during development, disease, and tissue regeneration. Phagocytes are responsible for clearing away dying cells and are typically characterized as either professional or nonprofessional phagocytes. Professional phagocytes, such as macrophages, are found in nearly every part of the body while nonprofessional phagocytes, such as epithelial cells, are found in every tissue type. However, there are organs that are considered "immune-privileged" as they have little to no immune surveillance and rely on nonprofessional phagocytes to engulf dying cells. These organs are surrounded by barriers to protect the tissue from viruses, bacteria, and perhaps even immune cells. The Drosophila ovary is considered immune-privileged, however the presence of hemocytes, the macrophages of Drosophila, around the ovary suggests they may have a potential function. Here we analyze hemocyte localization and potential functions in response to starvation-induced cell death in the ovary. Hemocytes were found to accumulate in the oviduct in the vicinity of mature eggs and follicle cell debris. Genetic ablation of hemocytes revealed that the presence of hemocytes affects oogenesis and that they phagocytose ovarian cell debris and in their absence fecundity decreases. Unpaired3, an IL-6 like cytokine, was found to be required for the recruitment of hemocytes to the oviduct to clear away obsolete follicle cells. These findings demonstrate a role for hemocytes in the ovary, providing a more thorough understanding of phagocyte communication and cell clearance in a previously thought immune-privileged organ.
Collapse
Affiliation(s)
- Alexandra Y. Chasse
- Program in Molecular Biology, Cell Biology & Biochemistry, Boston University, Boston, MA, United States
| | - Shruthi Bandyadka
- Program in Bioinformatics, Boston University, Boston, MA, United States
| | | | - Sandy B. Serizier
- Program in Molecular Biology, Cell Biology & Biochemistry, Boston University, Boston, MA, United States
| | - Kimberly McCall
- Department of Biology, Boston University, Boston, MA, United States
| |
Collapse
|
6
|
Martini L, Baek SH, Lo I, Raby BA, Silverman E, Weiss S, Glass K, Halu A. Detecting and dissecting signaling crosstalk via the multilayer network integration of signaling and regulatory interactions. Nucleic Acids Res 2024; 52:e5. [PMID: 37953325 PMCID: PMC10783515 DOI: 10.1093/nar/gkad1035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 06/27/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023] Open
Abstract
The versatility of cellular response arises from the communication, or crosstalk, of signaling pathways in a complex network of signaling and transcriptional regulatory interactions. Understanding the various mechanisms underlying crosstalk on a global scale requires untargeted computational approaches. We present a network-based statistical approach, MuXTalk, that uses high-dimensional edges called multilinks to model the unique ways in which signaling and regulatory interactions can interface. We demonstrate that the signaling-regulatory interface is located primarily in the intermediary region between signaling pathways where crosstalk occurs, and that multilinks can differentiate between distinct signaling-transcriptional mechanisms. Using statistically over-represented multilinks as proxies of crosstalk, we infer crosstalk among 60 signaling pathways, expanding currently available crosstalk databases by more than five-fold. MuXTalk surpasses existing methods in terms of model performance metrics, identifies additions to manual curation efforts, and pinpoints potential mediators of crosstalk. Moreover, it accommodates the inherent context-dependence of crosstalk, allowing future applications to cell type- and disease-specific crosstalk.
Collapse
Affiliation(s)
- Leonardo Martini
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Computer, Control, and Management Engineering, Sapienza University of Rome, Rome, 00185, Italy
| | - Seung Han Baek
- Division of Pulmonary Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ian Lo
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Benjamin A Raby
- Division of Pulmonary Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Arda Halu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
7
|
Klumpe HE, Garcia-Ojalvo J, Elowitz MB, Antebi YE. The computational capabilities of many-to-many protein interaction networks. Cell Syst 2023; 14:430-446. [PMID: 37348461 PMCID: PMC10318606 DOI: 10.1016/j.cels.2023.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/14/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023]
Abstract
Many biological circuits comprise sets of protein variants that interact with one another in a many-to-many, or promiscuous, fashion. These architectures can provide powerful computational capabilities that are especially critical in multicellular organisms. Understanding the principles of biochemical computations in these circuits could allow more precise control of cellular behaviors. However, these systems are inherently difficult to analyze, due to their large number of interacting molecular components, partial redundancies, and cell context dependence. Here, we discuss recent experimental and theoretical advances that are beginning to reveal how promiscuous circuits compute, what roles those computations play in natural biological contexts, and how promiscuous architectures can be applied for the design of synthetic multicellular behaviors.
Collapse
Affiliation(s)
- Heidi E Klumpe
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Jordi Garcia-Ojalvo
- Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain.
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Yaron E Antebi
- Department of Molecular Genetics, Weizmann Institute of Science 76100, Rehovot, Israel.
| |
Collapse
|
8
|
Long Q, Sun MH, Fan XX, Cai ZB, Zhang KY, Wang SY, Zhang JX, Gu XY, Song YX, Chen DF, Fu ZM, Guo R, Niu QS. First Identification and Investigation of piRNAs in the Larval Gut of the Asian Honeybee, Apis cerana. INSECTS 2022; 14:insects14010016. [PMID: 36661944 PMCID: PMC9863445 DOI: 10.3390/insects14010016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 05/31/2023]
Abstract
Piwi-interacting RNAs (piRNAs), a class of small non-coding RNAs (ncRNAs), play pivotal roles in maintaining the genomic stability and modulating biological processes such as growth and development via the regulation of gene expression. However, the piRNAs in the Asian honeybee (Apis cerana) are still largely unknown at present. In this current work, on the basis of previously gained high-quality small RNA-seq datasets, piRNAs in the larval gut of Apis cerana cerana, the nominated species of A. cerana, were identified for the first time, followed by an in-depth investigation of the regulatory roles of differentially expressed piRNAs (DEpiRNAs) in the developmental process of the A. c. cerana. Here, a total of 621 piRNAs were identified in A. c. cerana larval guts, among which 499 piRNAs were shared by 4-(Ac4 group), 5-(Ac5 group), and 6-day-old (Ac6 group) larval guts, while the numbers of unique ones equaled 79, 37, and 11, respectively. The piRNAs in each group ranged from 24 nucleotides (nt) to 33 nt in length, and the first base of the piRNAs had a cytosine (C) bias. Additionally, five up-regulated and five down-regulated piRNAs were identified in the Ac4 vs. Ac5 comparison group, nine of which could target 9011 mRNAs; these targets were involved in 41 GO terms and 137 pathways. Comparatively, 22 up-regulated piRNAs were detected in the Ac5 vs. Ac6 comparison group, 21 of which could target 28,969 mRNAs; these targets were engaged in 46 functional terms and 164 pathways. The results suggested an overall alteration of the expression pattern of piRNAs during the developmental process of A. c. cerana larvae. The regulatory network analysis showed that piR-ace-748815 and piR-ace-512574 in the Ac4 vs. Ac5 comparison group as well as piR-ace-716466 and piR-ace-828146 in the Ac5 vs. Ac6 comparison group were linked to the highest number of targets. Further investigation indicated that targets of DEpiRNAs in the abovementioned two comparison groups could be annotated to several growth and development-associated pathways, such as the Jak/STAT, TGF-β, and Wnt signaling pathways, indicating the involvement of DEpiRNAs in modulating larval gut development via these crucial pathways. Moreover, the expression trends of six randomly selected DEpiRNAs were verified using a combination of stem-loop RT-PCR and RT-qPCR. These results not only provide a novel insight into the development of the A. c. cerana larval gut, but also lay a foundation for uncovering the epigenetic mechanism underlying larval gut development.
Collapse
Affiliation(s)
- Qi Long
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ming-Hui Sun
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiao-Xue Fan
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zong-Bing Cai
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Kai-Yao Zhang
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Si-Yi Wang
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jia-Xin Zhang
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiao-Yu Gu
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yu-Xuan Song
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Da-Fu Chen
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Jilin Institute of Apicultural Research, Jilin 132000, China
| | - Zhong-Min Fu
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Jilin Institute of Apicultural Research, Jilin 132000, China
| | - Rui Guo
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Jilin Institute of Apicultural Research, Jilin 132000, China
| | - Qing-Sheng Niu
- Jilin Institute of Apicultural Research, Jilin 132000, China
- Apitherapy Research Institute, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
9
|
Dong Y, Dong S, Dizaji NB, Rutkowski N, Pohlenz T, Myles K, Dimopoulos G. The Aedes aegypti siRNA pathway mediates broad-spectrum defense against human pathogenic viruses and modulates antibacterial and antifungal defenses. PLoS Biol 2022; 20:e3001668. [PMID: 35679279 PMCID: PMC9182253 DOI: 10.1371/journal.pbio.3001668] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 05/11/2022] [Indexed: 01/08/2023] Open
Abstract
The mosquito's innate immune system defends against a variety of pathogens, and the conserved siRNA pathway plays a central role in the control of viral infections. Here, we show that transgenic overexpression of Dicer2 (Dcr2) or R2d2 resulted in an accumulation of 21-nucleotide viral sequences that was accompanied by a significant suppression of dengue virus (DENV), Zika virus (ZIKV), and chikungunya virus (CHIKV) replication, thus indicating the broad-spectrum antiviral response mediated by the siRNA pathway that can be applied for the development of novel arbovirus control strategies. Interestingly, overexpression of Dcr2 or R2d2 regulated the mRNA abundance of a variety of antimicrobial immune genes, pointing to additional functions of DCR2 and R2D2 as well as cross-talk between the siRNA pathway and other immune pathways. Accordingly, transgenic overexpression of Dcr2 or R2d2 resulted in a lesser proliferation of the midgut microbiota and increased resistance to bacterial and fungal infections.
Collapse
Affiliation(s)
- Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Nahid Borhani Dizaji
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Natalie Rutkowski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Tyler Pohlenz
- Texas A & M University, Department of Entomology, TAMU College Station, Texas, United States of America
| | - Kevin Myles
- Texas A & M University, Department of Entomology, TAMU College Station, Texas, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
10
|
Knaden M, Anderson P, Andersson MN, Hill SR, Sachse S, Sandgren M, Stensmyr MC, Löfstedt C, Ignell R, Hansson BS. Human Impacts on Insect Chemical Communication in the Anthropocene. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.791345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The planet is presently undergoing dramatic changes caused by human activities. We are living in the era of the Anthropocene, where our activities directly affect all living organisms on Earth. Insects constitute a major part of the world’s biodiversity and currently, we see dwindling insect biomass but also outbreaks of certain populations. Most insects rely on chemical communication to locate food, mates, and suitable oviposition sites, but also to avoid enemies and detrimental microbes. Emissions of, e.g., CO2, NOx, and ozone can all affect the chemical communication channel, as can a rising temperature. Here, we present a review of the present state of the art in the context of anthropogenic impact on insect chemical communication. We concentrate on present knowledge regarding fruit flies, mosquitoes, moths, and bark beetles, as well as presenting our views on future developments and needs in this emerging field of research. We include insights from chemical, physiological, ethological, and ecological directions and we briefly present a new international research project, the Max Planck Centre for Next Generation Insect Chemical Ecology (nGICE), launched to further increase our understanding of the impact of human activities on insect olfaction and chemical communication.
Collapse
|
11
|
Trapotsi MA, Hosseini-Gerami L, Bender A. Computational analyses of mechanism of action (MoA): data, methods and integration. RSC Chem Biol 2022; 3:170-200. [PMID: 35360890 PMCID: PMC8827085 DOI: 10.1039/d1cb00069a] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022] Open
Abstract
The elucidation of a compound's Mechanism of Action (MoA) is a challenging task in the drug discovery process, but it is important in order to rationalise phenotypic findings and to anticipate potential side-effects. Bioinformatic approaches, advances in machine learning techniques and the increasing deposition of high-throughput data in public databases have significantly contributed to recent advances in the field, but it is not straightforward to decide which data and methods are most suitable to use in a given case. In this review, we focus on these methods and data and their applications in generating MoA hypotheses for subsequent experimental validation. We discuss compound-specific data such as -omics, cell morphology and bioactivity data, as well as commonly used supplementary prior knowledge such as network and pathway data, and provide information on databases where this data can be accessed. In terms of methodologies, we discuss both well-established methods (connectivity mapping, pathway enrichment) as well as more developing methods (neural networks and multi-omics integration). Finally, we review case studies where the MoA of a compound was successfully suggested from computational analysis by incorporating multiple data modalities and/or methodologies. Our aim for this review is to provide researchers with insights into the benefits and drawbacks of both the data and methods in terms of level of understanding, biases and interpretation - and to highlight future avenues of investigation which we foresee will improve the field of MoA elucidation, including greater public access to -omics data and methodologies which are capable of data integration.
Collapse
Affiliation(s)
- Maria-Anna Trapotsi
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| | - Layla Hosseini-Gerami
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| | - Andreas Bender
- Centre for Molecular Informatics, Yusuf Hamied Department of Chemistry, University of Cambridge UK
| |
Collapse
|
12
|
|
13
|
Novel doxorubicin / folate-targeted trans-ferulic acid-loaded PLGA nanoparticles combination: In-vivo superiority over standard chemotherapeutic regimen for breast cancer treatment. Biomed Pharmacother 2021; 145:112376. [PMID: 34749055 DOI: 10.1016/j.biopha.2021.112376] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/16/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
AIM Doxorubicin/Cyclophosphamide (AC) is one of the standard adjuvant anthracycline-containing regimens that is still in use for breast cancer treatment. Cancer cell resistance and AC-induced side effects make treatment suboptimal and worsen patients' quality of life. This study aimed to improve trans-ferulic acid's (TFA) efficiency via loading into folate-receptor-targeted-poly lactic-co-glycolic acid nanoparticles (FA-PLGA-TFA NPs). Also, investigating both the antitumor efficacy of Doxorubicin (Dox)/FA-PLGA-TFA NPs combination against dimethylbenz[a]anthracene (DMBA)-induced breast cancer and its safety profile. METHODS FA-PLGA-TFA NPs were optimally fabricated and characterized. Levels of Notch1, Hes1, Wnt-3a, β-catenin, MMP-9, cyclin D1, Permeability-Glycoprotein (P-gp), ERα, PR, and HER2 were assessed as a measure of the antitumor efficacy of different treatment protocols. Histopathological examination of heart and bone, levels of ALT, AST, ALP, CK-MB, and WBCs count were evaluated to ensure the combination's safety profile. KEY FINDINGS Dox/FA-PLGA-TFA NPs not only inhibited Notch signaling but also suppressed Notch synergy with Wnt, estrogen, progesterone, and HER2 pathways. Interestingly, Dox/FA-PLGA-TFA NPs decreased P-gp level and preserved heart, bone, and liver health as well as WBCs count. SIGNIFICANCE Dox/FA-PLGA-TFA NPs reduced the side-effects of each single drug, and at the same time exerted excellent antitumor activity that surpass the AC regimen in evading cancer cell resistance and having a superior safety profile.
Collapse
|
14
|
Wang L, Sun X, He J, Liu Z. Functions and Molecular Mechanisms of Deltex Family Ubiquitin E3 Ligases in Development and Disease. Front Cell Dev Biol 2021; 9:706997. [PMID: 34513839 PMCID: PMC8424196 DOI: 10.3389/fcell.2021.706997] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Ubiquitination is a posttranslational modification of proteins that significantly affects protein stability and function. The specificity of substrate recognition is determined by ubiquitin E3 ligase during ubiquitination. Human Deltex (DTX) protein family, which functions as ubiquitin E3 ligases, comprises five members, namely, DTX1, DTX2, DTX3, DTX3L, and DTX4. The characteristics and functional diversity of the DTX family proteins have attracted significant attention over the last decade. DTX proteins have several physiological and pathological roles and are closely associated with cell signal transduction, growth, differentiation, and apoptosis, as well as the occurrence and development of various tumors. Although they have been extensively studied in various species, data on structural features, biological functions, and potential mechanisms of action of the DTX family proteins remain limited. In this review, recent research progress on each member of the DTX family is summarized, providing insights into future research directions and potential strategies in disease diagnosis and therapy.
Collapse
Affiliation(s)
- Lidong Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodan Sun
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Jingni He
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Cheng J, Zhang J, Wu Z, Sun X. Inferring microenvironmental regulation of gene expression from single-cell RNA sequencing data using scMLnet with an application to COVID-19. Brief Bioinform 2021; 22:988-1005. [PMID: 33341869 PMCID: PMC7799217 DOI: 10.1093/bib/bbaa327] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/18/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Abstract
Inferring how gene expression in a cell is influenced by cellular microenvironment is of great importance yet challenging. In this study, we present a single-cell RNA-sequencing data based multilayer network method (scMLnet) that models not only functional intercellular communications but also intracellular gene regulatory networks (https://github.com/SunXQlab/scMLnet). scMLnet was applied to a scRNA-seq dataset of COVID-19 patients to decipher the microenvironmental regulation of expression of SARS-CoV-2 receptor ACE2 that has been reported to be correlated with inflammatory cytokines and COVID-19 severity. The predicted elevation of ACE2 by extracellular cytokines EGF, IFN-γ or TNF-α were experimentally validated in human lung cells and the related signaling pathway were verified to be significantly activated during SARS-COV-2 infection. Our study provided a new approach to uncover inter-/intra-cellular signaling mechanisms of gene expression and revealed microenvironmental regulators of ACE2 expression, which may facilitate designing anti-cytokine therapies or targeted therapies for controlling COVID-19 infection. In addition, we summarized and compared different methods of scRNA-seq based inter-/intra-cellular signaling network inference for facilitating new methodology development and applications.
Collapse
Affiliation(s)
- Jinyu Cheng
- Zhong-Shan School of Medicine, Sun Yat-Sen University
| | - Ji Zhang
- Department of Neurosurgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center
| | - Zhongdao Wu
- Zhong-Shan School of Medicine, Sun Yat-Sen University
| | - Xiaoqiang Sun
- Zhong-Shan School of Medicine, Sun Yat-Sen University
| |
Collapse
|
16
|
Cambron LD, Yocum GD, Yeater KM, Greenlee KJ. Overwintering conditions impact insulin pathway gene expression in diapausing Megachile rotundata. Comp Biochem Physiol A Mol Integr Physiol 2021; 256:110937. [PMID: 33737040 DOI: 10.1016/j.cbpa.2021.110937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/22/2022]
Abstract
Diapause is a non-feeding state that many insects undergo to survive the winter months. With fixed resources, overall metabolism and insulin signaling (IIS) are maintained at low levels, but whether those change in response to seasonal temperature fluctuations remains unknown. The focus of this study was to determine 1) how genes in the insulin signaling pathway vary throughout diapause and 2) if that variation changes in response to temperature. To test the hypothesis that expression of IIS pathway genes vary in response to temperature fluctuations during overwintering, alfalfa leafcutting bees, Megachile rotundata, were overwintered at either a constant 4 °C in the lab or in naturally fluctuating temperatures in the field. Expression levels of genes in the IIS pathway, cell cycle regulators, and transcription factors were measured. Overall our findings showed that a few key targets of the insulin signaling pathway, along with growth regulators, change during overwintering, suggesting that only cell cycle regulators, and not the IIS pathway as a whole, change across the phases of diapause. To answer our second question, we compared gene expression levels between temperature treatments at each month for a given gene. We observed significantly more differences in expression of IIS pathway targets, indicating that overwintering conditions impact insulin pathway gene expression and leads to altered expression profiles. With differences seen between temperature treatment groups, these findings indicate that constant temperatures like those used in agricultural storage protocols, lead to different expression profiles and possibly different diapause phenotypes for alfalfa leafcutting bees.
Collapse
Affiliation(s)
| | | | - Kathleen M Yeater
- USDA-ARS, Plains Area Office of the Director, Fort Collins, CO, United States of America
| | | |
Collapse
|
17
|
AlMusawi S, Ahmed M, Nateri AS. Understanding cell-cell communication and signaling in the colorectal cancer microenvironment. Clin Transl Med 2021; 11:e308. [PMID: 33635003 PMCID: PMC7868082 DOI: 10.1002/ctm2.308] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/31/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Carcinomas are complex heterocellular systems containing epithelial cancer cells, stromal fibroblasts, and multiple immune cell-types. Cell-cell communication between these tumor microenvironments (TME) and cells drives cancer progression and influences response to existing therapies. In order to provide better treatments for patients, we must understand how various cell-types collaborate within the TME to drive cancer and consider the multiple signals present between and within different cancer types. To investigate how tissues function, we need a model to measure both how signals are transferred between cells and how that information is processed within cells. The interplay of collaboration between different cell-types requires cell-cell communication. This article aims to review the current in vitro and in vivo mono-cellular and multi-cellular cultures models of colorectal cancer (CRC), and to explore how they can be used for single-cell multi-omics approaches for isolating multiple types of molecules from a single-cell required for cell-cell communication to distinguish cancer cells from normal cells. Integrating the existing single-cell signaling measurements and models, and through understanding the cell identity and how different cell types communicate, will help predict drug sensitivities in tumor cells and between- and within-patients responses.
Collapse
Affiliation(s)
- Shaikha AlMusawi
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Division of Cancer & Stem Cells, School of MedicineUniversity of NottinghamNottinghamUK
| | - Mehreen Ahmed
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Division of Cancer & Stem Cells, School of MedicineUniversity of NottinghamNottinghamUK
- Department of Laboratory Medicine, Division of Translational Cancer ResearchLund UniversityLundSweden
| | - Abdolrahman S. Nateri
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Division of Cancer & Stem Cells, School of MedicineUniversity of NottinghamNottinghamUK
| |
Collapse
|
18
|
Krautz R, Khalili D, Theopold U. Tissue-autonomous immune response regulates stress signaling during hypertrophy. eLife 2020; 9:64919. [PMID: 33377870 PMCID: PMC7880693 DOI: 10.7554/elife.64919] [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: 11/15/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Postmitotic tissues are incapable of replacing damaged cells through proliferation, but need to rely on buffering mechanisms to prevent tissue disintegration. By constitutively activating the Ras/MAPK-pathway via RasV12-overexpression in the postmitotic salivary glands (SGs) of Drosophila larvae, we overrode the glands adaptability to growth signals and induced hypertrophy. The accompanied loss of tissue integrity, recognition by cellular immunity, and cell death are all buffered by blocking stress signaling through a genuine tissue-autonomous immune response. This novel, spatio-temporally tightly regulated mechanism relies on the inhibition of a feedback-loop in the JNK-pathway by the immune effector and antimicrobial peptide Drosomycin. While this interaction might allow growing SGs to cope with temporary stress, continuous Drosomycin expression in RasV12-glands favors unrestricted hypertrophy. These findings indicate the necessity to refine therapeutic approaches that stimulate immune responses by acknowledging their possible, detrimental effects in damaged or stressed tissues. Tissues and organs work hard to maintain balance in everything from taking up nutrients to controlling their growth. Ageing, wounding, sickness, and changes in the genetic code can all alter this balance, and cause the tissue or organ to lose some of its cells. Many tissues restore this loss by dividing their remaining cells to fill in the gaps. But some – like the salivary glands of fruit fly larvae – have lost this ability. Tissues like these rely on being able to sense and counteract problems as they arise so as to not lose their balance in the first place. The immune system and stress responses are crucial for this process. They trigger steps to correct the problem and interact with each other to find a common decision about the fate of the affected tissue. To better understand how the immune system and stress response work together, Krautz, Khalili and Theopold genetically manipulated cells in the salivary gland of fruit fly larvae. These modifications switched on signals that stimulate cells to keep growing, causing the salivary gland’s tissue to slowly lose its balance and trigger the stress and immune response. The experiments showed that while the stress response instructed the cells in the gland to die, a peptide released by the immune system called Drosomycin blocked this response and prevented the tissue from collapsing. The cells in the part of the gland not producing this immune peptide were consequently killed by the stress response. When all the cells in the salivary gland were forced to produce Drosomycin, none of the cells died and the whole tissue survived. But it also allowed the cells in the gland to grow uncontrollably, like a tumor, threatening the health of the entire organism. Mapping the interactions between immune and stress pathways could help to fine-tune treatments that can prevent tissue damage. Fruit flies share many genetic features and molecular pathways with humans. So, the next step towards these kinds of treatments would be to screen for similar mechanisms that block stress activation in damaged human tissues. But this research carries a warning: careless activation of the immune system to protect stressed tissues could lead to uncontrolled tissue growth, and might cause more harm than good.
Collapse
Affiliation(s)
- Robert Krautz
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Dilan Khalili
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Ulrich Theopold
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| |
Collapse
|
19
|
Hongdusit A, Liechty ET, Fox JM. Optogenetic interrogation and control of cell signaling. Curr Opin Biotechnol 2020; 66:195-206. [PMID: 33053496 DOI: 10.1016/j.copbio.2020.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 02/05/2023]
Abstract
Signaling networks control the flow of information through biological systems and coordinate the chemical processes that constitute cellular life. Optogenetic actuators - genetically encoded proteins that undergo light-induced changes in activity or conformation - are useful tools for probing signaling networks over time and space. They have permitted detailed dissections of cellular proliferation, differentiation, motility, and death, and enabled the assembly of synthetic systems with applications in areas as diverse as photography, chemical synthesis, and medicine. In this review, we provide a brief introduction to optogenetic systems and describe their application to molecular-level analyses of cell signaling. Our discussion highlights important research achievements and speculates on future opportunities to exploit optogenetic systems in the study and assembly of complex biochemical networks.
Collapse
Affiliation(s)
- Akarawin Hongdusit
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80303, USA
| | - Evan T Liechty
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80303, USA
| | - Jerome M Fox
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80303, USA.
| |
Collapse
|
20
|
Towarnicki SG, Ballard JWO. Towards understanding the evolutionary dynamics of mtDNA. Mitochondrial DNA A DNA Mapp Seq Anal 2020; 31:355-364. [PMID: 33026269 DOI: 10.1080/24701394.2020.1830076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Historically, mtDNA was considered a selectively neutral marker that was useful for estimating the population genetic history of the maternal lineage. Over time there has been an increasing appreciation of mtDNA and mitochondria in maintaining cellular and organismal health. Beyond energy production, mtDNA and mitochondria have critical cellular roles in signalling. Here we briefly review the structure of mtDNA and the role of the mitochondrion in energy production. We then discuss the predictions that can be obtained from quaternary structure modelling and focus on mitochondrial complex I. Complex I is the primary entry point for electrons into the electron transport system is the largest respiratory complex of the chain and produces about 40% of the proton flux used to synthesize ATP. A focus of the review is Drosophila's utility as a model organism to study the selective advantage of specific mutations. However, we note that the incorporation of insights from a multitude of systems is necessary to fully understand the range of roles that mtDNA has in organismal fitness. We speculate that dietary changes can illicit stress responses that influence the selective advantage of specific mtDNA mutations and cause spatial and temporal fluctuations in the frequencies of mutations. We conclude that developing our understanding of the roles mtDNA has in determining organismal fitness will enable increased evolutionary insight and propose we can no longer assume it is evolving as a strictly neutral marker without testing this hypothesis.
Collapse
Affiliation(s)
- Samuel G Towarnicki
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| | - J William O Ballard
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| |
Collapse
|
21
|
Kuo A, Magiera J, Rethwan N, Andersson Å, Leen Lam A, Wyse B, Meutermans W, Lewis R, Smith M. In vitro profiling of opioid ligands using the cAMP formation inhibition assay and the β-arrestin2 recruitment assay: No two ligands have the same profile. Eur J Pharmacol 2020; 872:172947. [PMID: 31991138 DOI: 10.1016/j.ejphar.2020.172947] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 10/25/2022]
Abstract
Previously, we showed that no two of seven opioids administered by the intracerebroventricular route had the same potency rank order for evoking antinociception, constipation and respiratory depression in rats. To gain insight at the cellular level, this study was designed to systematically investigate the activity profiles of six commonly used opioid ligands using the forskolin-stimulated cAMP assay and a β-arrestin2 recruitment assay in cultured HEK-293 cells transfected with MOP(μ), DOP(δ) or KOP(κ) receptors(-r). Morphine was a potent agonist at the MOP-r in the cAMP assay whereas it was a weak agonist at the KOP-r and DOP-r. Oxycodone had moderate efficacy and low potency at the MOP-r. Buprenorphine was a potent MOP-r and DOP-r agonist; its efficacy rank order was DOP > MOP > KOP. Fentanyl was a potent agonist at the MOP-r; its efficacy rank order was MOP > DOP > KOP. For DPDPE, its agonist efficacy was confined to the DOP-r, whereas for U69593, its efficacy rank order was KOP>> MOP. For the β-arrestin2 assay, fentanyl had full efficacy at the MOP-r whereas morphine and oxycodone were weak with insignificant efficacy at DOP and KOP receptors. Buprenorphine did not recruit β-arrestin2 at all three opioid-receptors. DPDPE and U69593 had full efficacy for β-arrestin2 recruitment to the DOP-r and KOP-r respectively. Despite the low efficacy and potency of morphine, oxycodone and buprenorphine in recruiting β-arrestin2 to the MOP-r herein, these opioids all evoked respiratory depression and constipation in rats. Together, our findings discount a key role for β-arrestin2 recruitment at the MOP-r in evoking opioid-related side-effects.
Collapse
Affiliation(s)
- Andy Kuo
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Julia Magiera
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Nursyazwani Rethwan
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Åsa Andersson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Ai Leen Lam
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Bruce Wyse
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Wim Meutermans
- VAST Bioscience Pty Ltd, Toowong, Brisbane, QLD, Australia
| | - Richard Lewis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Maree Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
22
|
Reddy D, Kumavath R, Ghosh P, Barh D. Lanatoside C Induces G2/M Cell Cycle Arrest and Suppresses Cancer Cell Growth by Attenuating MAPK, Wnt, JAK-STAT, and PI3K/AKT/mTOR Signaling Pathways. Biomolecules 2019; 9:biom9120792. [PMID: 31783627 PMCID: PMC6995510 DOI: 10.3390/biom9120792] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 01/09/2023] Open
Abstract
Cardiac glycosides (CGs) are a diverse family of naturally derived compounds having a steroid and glycone moiety in their structures. CG molecules inhibit the α-subunit of ubiquitous transmembrane protein Na+/K+-ATPase and are clinically approved for the treatment of cardiovascular diseases. Recently, the CGs were found to exhibit selective cytotoxic effects against cancer cells, raising interest in their use as anti-cancer molecules. In this current study, we explored the underlying mechanism responsible for the anti-cancer activity of Lanatoside C against breast (MCF-7), lung (A549), and liver (HepG2) cancer cell lines. Using Real-time PCR, western blot, and immunofluorescence studies, we observed that (i) Lanatoside C inhibited cell proliferation and induced apoptosis in cell-specific and dose-dependent manner only in cancer cell lines; (ii) Lanatoside C exerts its anti-cancer activity by arresting the G2/M phase of cell cycle by blocking MAPK/Wnt/PAM signaling pathways; (iii) it induces apoptosis by inducing DNA damage and inhibiting PI3K/AKT/mTOR signaling pathways; and finally, (iv) molecular docking analysis shows significant evidence on the binding sites of Lanatoside C with various key signaling proteins ranging from cell survival to cell death. Our studies provide a novel molecular insight of anti-cancer activities of Lanatoside C in human cancer cells.
Collapse
Affiliation(s)
- Dhanasekhar Reddy
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (P.O) Kasaragod 671316, Kerala, India;
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (P.O) Kasaragod 671316, Kerala, India;
- Correspondence: or ; Tel.: +91-8547-648-620
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, West Bengal, India;
| |
Collapse
|
23
|
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
|
24
|
La Marca JE, Diepstraten ST, Hodge AL, Wang H, Hart AH, Richardson HE, Somers WG. Strip and Cka negatively regulate JNK signalling during Drosophila spermatogenesis. Development 2019; 146:dev.174292. [PMID: 31164352 DOI: 10.1242/dev.174292] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 05/24/2019] [Indexed: 12/16/2022]
Abstract
One fundamental property of a stem cell niche is the exchange of molecular signals between its component cells. Niche models, such as the Drosophila melanogaster testis, have been instrumental in identifying and studying the conserved genetic factors that contribute to niche molecular signalling. Here, we identify jam packed (jam), an allele of Striatin interacting protein (Strip), which is a core member of the highly conserved Striatin-interacting phosphatase and kinase (STRIPAK) complex. In the developing Drosophila testis, Strip cell-autonomously regulates the differentiation and morphology of the somatic lineage, and non-cell-autonomously regulates the proliferation and differentiation of the germline lineage. Mechanistically, Strip acts in the somatic lineage with its STRIPAK partner, Connector of kinase to AP-1 (Cka), where they negatively regulate the Jun N-terminal kinase (JNK) signalling pathway. Our study reveals a novel role for Strip/Cka in JNK pathway regulation during spermatogenesis within the developing Drosophila testis.
Collapse
Affiliation(s)
- John E La Marca
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia 3086
| | - Sarah T Diepstraten
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia 3086
| | - Amy L Hodge
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia 3086
| | - Hongyan Wang
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857.,National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Adam H Hart
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia 3086
| | - Helena E Richardson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia 3086
| | - W Gregory Somers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia 3086
| |
Collapse
|
25
|
Zhang J, Guan M, Wang Q, Zhang J, Zhou T, Sun X. Single-cell transcriptome-based multilayer network biomarker for predicting prognosis and therapeutic response of gliomas. Brief Bioinform 2019; 21:1080-1097. [DOI: 10.1093/bib/bbz040] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/22/2019] [Accepted: 03/12/2019] [Indexed: 12/23/2022] Open
Abstract
Abstract
Occurrence and development of cancers are governed by complex networks of interacting intercellular and intracellular signals. The technology of single-cell RNA sequencing (scRNA-seq) provides an unprecedented opportunity for dissecting the interplay between the cancer cells and the associated microenvironment. Here we combined scRNA-seq data with clinical bulk gene expression data to develop a computational pipeline for identifying the prognostic and predictive signature that connects cancer cells and microenvironmental cells. The pipeline was applied to glioma scRNA-seq data and revealed a tumor-associated microglia/macrophage-mediated EGFR/ERBB2 feedback-crosstalk signaling module, which was defined as a multilayer network biomarker (MNB) to predict survival outcome and therapeutic response of glioma patients. We used publicly available clinical data sets from large cohorts of glioma patients to examine the prognostic significance and predictive accuracy of the MNB, which outperformed conventional gene biomarkers and other methods. Additionally, the MNB was found to be predictive of the sensitivity or resistance of glioma patients to molecularly targeted therapeutics. Moreover, the MNB was an independent and the strongest prognostic factor when adjusted for clinicopathologic risk factors and other existing gene signatures. The robustness of the MNB was further tested on additional data sets. Our study presents a promising scRNA-seq transcriptome-based multilayer network approach to elucidate the interactions between tumor cell and tumor-associated microenvironment and to identify prognostic and predictive signatures of cancer patients. The proposed MNB method may facilitate the design of more effective biomarkers for predicting prognosis and therapeutic resistance of cancer patients.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Neurosurgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Meige Guan
- School of Mathematics, Sun Yat-Sen University, Guangzhou, China
| | - Qianliang Wang
- School of Life Science, Sun Yat-Sen University, Guangzhou, China
| | - Jiajun Zhang
- School of Mathematics, Sun Yat-Sen University, Guangzhou, China
| | - Tianshou Zhou
- School of Mathematics, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoqiang Sun
- Department of Medical Informatics, Zhong-shan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China
- School of Mathematics, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
26
|
Neal SJ, Dolezal D, Jusić N, Pignoni F. Drosophila ML-DmD17-c3 cells respond robustly to Dpp and exhibit complex transcriptional feedback on BMP signaling components. BMC DEVELOPMENTAL BIOLOGY 2019; 19:1. [PMID: 30669963 PMCID: PMC6341649 DOI: 10.1186/s12861-019-0181-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/07/2019] [Indexed: 01/07/2023]
Abstract
Background BMP signaling is involved in myriad metazoan developmental processes, and study of this pathway in Drosophila has contributed greatly to our understanding of its molecular and genetic mechanisms. These studies have benefited not only from Drosophila’s advanced genetic tools, but from complimentary in vitro culture systems. However, the commonly-used S2 cell line is not intrinsically sensitive to the major BMP ligand Dpp and must therefore be augmented with exogenous pathway components for most experiments. Results Herein we identify and characterize the responses of Drosophila ML-DmD17-c3 cells, which are sensitive to Dpp stimulation and exhibit characteristic regulation of BMP target genes including Dad and brk. Dpp signaling in ML-DmD17-c3 cells is primarily mediated by the receptors Put and Tkv, with additional contributions from Wit and Sax. Furthermore, we report complex regulatory feedback on core pathway genes in this system. Conclusions Native ML-DmD17-c3 cells exhibit robust transcriptional responses to BMP pathway induction. We propose that ML-DmD17-c3 cells are well-suited for future BMP pathway analyses. Electronic supplementary material The online version of this article (10.1186/s12861-019-0181-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Scott J Neal
- Center for Vision Research and Department of Ophthalmology, Upstate Medical University, NRB-4610, 505 Irving Ave, Syracuse, 13210, NY, USA.
| | - Darin Dolezal
- Center for Vision Research and Department of Ophthalmology, Upstate Medical University, NRB-4610, 505 Irving Ave, Syracuse, 13210, NY, USA.,Department of Biochemistry and Molecular Biology, Upstate Medical University, Syracuse, NY, USA.,Current Address: Department of Surgical Pathology, Yale-New Haven Hospital, New Haven, CT, USA
| | - Nisveta Jusić
- Center for Vision Research and Department of Ophthalmology, Upstate Medical University, NRB-4610, 505 Irving Ave, Syracuse, 13210, NY, USA
| | - Francesca Pignoni
- Center for Vision Research and Department of Ophthalmology, Upstate Medical University, NRB-4610, 505 Irving Ave, Syracuse, 13210, NY, USA. .,Department of Biochemistry and Molecular Biology, Upstate Medical University, Syracuse, NY, USA. .,Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
27
|
Luhur A, Klueg KM, Zelhof AC. Generating and working with Drosophila cell cultures: Current challenges and opportunities. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 8:e339. [PMID: 30561900 DOI: 10.1002/wdev.339] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/30/2018] [Accepted: 11/21/2018] [Indexed: 12/26/2022]
Abstract
The use of Drosophila cell cultures has positively impacted both fundamental and biomedical research. The most widely used cell lines: Schneider, Kc, the CNS and imaginal disc lines continue to be the choice for many applications. Drosophila cell lines provide a homogenous source of cells suitable for biochemical experimentations, transcriptomics, functional genomics, and biomedical applications. They are amenable to RNA interference and serve as a platform for high-throughput screens to identify relevant candidate genes or drugs for any biological process. Currently, CRISPR-based functional genomics are also being developed for Drosophila cell lines. Even though many uniquely derived cell lines exist, cell genetic techniques such the transgenic UAS-GAL4-based RasV12 oncogene expression, CRISPR-Cas9 editing and recombination mediated cassette exchange are likely to drive the establishment of many more lines from specific tissues, cells, or genotypes. However, the pace of creating new lines is hindered by several factors inherent to working with Drosophila cell cultures: single cell cloning, optimal media formulations and culture conditions capable of supporting lines from novel tissue sources or genotypes. Moreover, even though many Drosophila cell lines are morphologically and transcriptionally distinct it may be necessary to implement a standard for Drosophila cell line authentication, ensuring the identity and purity of each cell line. Altogether, recent advances and a standardized authentication effort should improve the utility of Drosophila cell cultures as a relevant model for fundamental and biomedical research. This article is categorized under: Technologies > Analysis of Cell, Tissue, and Animal Phenotypes.
Collapse
Affiliation(s)
- Arthur Luhur
- Department of Biology, Drosophila Genomics Resource Center, Indiana University Bloomington, Bloomington, Indiana
| | - Kristin M Klueg
- Department of Biology, Drosophila Genomics Resource Center, Indiana University Bloomington, Bloomington, Indiana
| | - Andrew C Zelhof
- Department of Biology, Drosophila Genomics Resource Center, Indiana University Bloomington, Bloomington, Indiana
| |
Collapse
|
28
|
Notch and Wnt Dysregulation and Its Relevance for Breast Cancer and Tumor Initiation. Biomedicines 2018; 6:biomedicines6040101. [PMID: 30388742 PMCID: PMC6315509 DOI: 10.3390/biomedicines6040101] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the second leading cause of cancer deaths among women in the world. Treatment has been improved and, in combination with early detection, this has resulted in reduced mortality rates. Further improvement in therapy development is however warranted. This will be particularly important for certain sub-classes of breast cancer, such as triple-negative breast cancer, where currently no specific therapies are available. An important therapy development focus emerges from the notion that dysregulation of two major signaling pathways, Notch and Wnt signaling, are major drivers for breast cancer development. In this review, we discuss recent insights into the Notch and Wnt signaling pathways and into how they act synergistically both in normal development and cancer. We also discuss how dysregulation of the two pathways contributes to breast cancer and strategies to develop novel breast cancer therapies starting from a Notch and Wnt dysregulation perspective.
Collapse
|
29
|
An efficient and multiple target transgenic RNAi technique with low toxicity in Drosophila. Nat Commun 2018; 9:4160. [PMID: 30297884 PMCID: PMC6175926 DOI: 10.1038/s41467-018-06537-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/12/2018] [Indexed: 01/19/2023] Open
Abstract
Being relatively simple and practical, Drosophila transgenic RNAi is the technique of top priority choice to quickly study genes with pleiotropic functions. However, drawbacks have emerged over time, such as high level of false positive and negative results. To overcome these shortcomings and increase efficiency, specificity and versatility, we develop a next generation transgenic RNAi system. With this system, the leaky expression of the basal promoter is significantly reduced, as well as the heterozygous ratio of transgenic RNAi flies. In addition, it has been first achieved to precisely and efficiently modulate highly expressed genes. Furthermore, we increase versatility which can simultaneously knock down multiple genes in one step. A case illustration is provided of how this system can be used to study the synthetic developmental effect of histone acetyltransferases. Finally, we have generated a collection of transgenic RNAi lines for those genes that are highly homologous to human disease genes.
Collapse
|
30
|
Impact of the microsporidian Nosema ceranae on the gut epithelium renewal of the honeybee, Apis mellifera. J Invertebr Pathol 2018; 159:121-128. [PMID: 30268675 DOI: 10.1016/j.jip.2018.09.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
The invasive microsporidian species, Nosema ceranae, causes nosemosis in honeybees and is suspected to be involved in Western honeybee (Apis mellifera) declines worldwide. The midgut of honeybees is the site of infection; the microsporidium can disturb the functioning of this organ and, thus, the bee physiology. Host defense against pathogens is not limited to resistance (i.e. the immune response) but also involves resilience. This process implies that the host can tolerate and repair damage inflicted by the infection- by the pathogen itself or by an excessive host immune response. Enterocyte damage caused by N. ceranae can be compensated by proliferation of intestinal stem cells (ISCs) that are under the control of multiple pathways. In the present study, we investigated the impact of N. ceranae on honeybee epithelium renewal by following the mitotic index of midgut stem cells during a 22-day N. ceranae infection. Fluorescence in situ hybridization (FISH) and immunostaining experiments were performed to follow the parasite proliferation/progression in the intestinal tissue, especially in the ISCs as they are key cells for the midgut homeostasis. We also monitored the transcriptomic profile of 7 genes coding for key proteins involved in pathways implicated in the gut epithelium renewal and homeostasis. We have shown for the first time that N. ceranae can negatively alter the gut epithelium renewal rate and disrupt some signaling pathways involved in the gut homeostasis. This alteration is correlated to a reduced longevity of N. ceranae-infected honeybees and we can assume that honeybee susceptibility to N. ceranae could be due to an impaired ability to repair gut damage.
Collapse
|
31
|
Wang H, Liu P, Li Q, Zhou T. Entangled signal pathways can both control expression stability and induce stochastic focusing. FEBS Lett 2018; 592:1135-1149. [DOI: 10.1002/1873-3468.13012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/28/2018] [Accepted: 02/08/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Haohua Wang
- Department of Mathematics College of Information Science and Technology Hainan University Haikou China
| | - Peijiang Liu
- School of Statistics and Mathematics Guangdong University of Finance & Economics Guangzhou China
| | - Qingqing Li
- Guangdong Province Key Laboratory of Computational Science School of Mathematics Sun Yat‐Sen University Guangzhou China
| | - Tianshou Zhou
- Guangdong Province Key Laboratory of Computational Science School of Mathematics Sun Yat‐Sen University Guangzhou China
| |
Collapse
|
32
|
Zheng L, Conner SD. Glycogen synthase kinase 3β inhibition enhances Notch1 recycling. Mol Biol Cell 2018; 29:389-395. [PMID: 29237816 PMCID: PMC6014177 DOI: 10.1091/mbc.e17-07-0474] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 11/11/2022] Open
Abstract
The Notch signaling pathway is essential throughout development and remains active into adulthood, where it performs a critical role in tissue homeostasis. The fact that defects in signaling can lead to malignancy illustrates the need to control Notch activity tightly. GSK3β is an established regulator of the Notch signaling pathway, although its mechanism of action remains unclear. Given the emerging role for GSK3β in receptor trafficking, we tested the idea that GSK3β controls signaling by regulating Notch transport. Consistent with published reports, we find that GSK3β inhibition enhances Notch1 signaling activity. Immunolocalization analysis reveals that Notch1 localization within a tubulovesicular compartment is altered when GSK3β activity is disrupted. We also find that receptor cell surface levels increase following acute GSK3β inhibition. This is followed by elevated Notch intra-cellular domain (NICD) production and a corresponding increase in signaling activity. Moreover, Notch transport assays reveal that receptor recycling rates increase when GSK3β activity is inhibited. Collectively, results presented here support a model where GSK3β regulates signaling by controlling postendocytic transport of Notch1. Given that GSK3β activity is suppressed following stimulation by multiple signal transduction pathways, our findings also suggest that cells can modulate Notch1 activity in response to extracellular signals by mobilizing Notch1 from endosomal stores.
Collapse
Affiliation(s)
- Li Zheng
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| | - Sean D Conner
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| |
Collapse
|
33
|
Ketosugbo KF, Bushnell HL, Johnson RI. A screen for E3 ubiquitination ligases that genetically interact with the adaptor protein Cindr during Drosophila eye patterning. PLoS One 2017; 12:e0187571. [PMID: 29117266 PMCID: PMC5678704 DOI: 10.1371/journal.pone.0187571] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/23/2017] [Indexed: 01/12/2023] Open
Abstract
Ubiquitination is a crucial post-translational modification that can target proteins for degradation. The E3 ubiquitin ligases are responsible for recognizing substrate proteins for ubiquitination, hence providing specificity to the process of protein degradation. Here, we describe a genetic modifier screen that identified E3 ligases that modified the rough-eye phenotype generated by expression of cindrRNAi transgenes during Drosophila eye development. In total, we identified 36 E3 ligases, as well as 4 Cullins, that modified the mild cindrRNA mis-patterning phenotype. This indicates possible roles for these E3s/Cullins in processes that require Cindr function, including cytoskeletal regulation, cell adhesion, cell signaling and cell survival. Three E3 ligases identified in our screen had previously been linked to regulating JNK signaling.
Collapse
Affiliation(s)
- Kwami F. Ketosugbo
- Biology Department, Wesleyan University, Middletown, Connecticut, United States of America
| | - Henry L. Bushnell
- Biology Department, Wesleyan University, Middletown, Connecticut, United States of America
| | - Ruth I. Johnson
- Biology Department, Wesleyan University, Middletown, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
34
|
Xu C, Luo J, He L, Montell C, Perrimon N. Oxidative stress induces stem cell proliferation via TRPA1/RyR-mediated Ca 2+ signaling in the Drosophila midgut. eLife 2017; 6. [PMID: 28561738 PMCID: PMC5451214 DOI: 10.7554/elife.22441] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 05/23/2017] [Indexed: 12/20/2022] Open
Abstract
Precise regulation of stem cell activity is crucial for tissue homeostasis and necessary to prevent overproliferation. In the Drosophila adult gut, high levels of reactive oxygen species (ROS) has been detected with different types of tissue damage, and oxidative stress has been shown to be both necessary and sufficient to trigger intestinal stem cell (ISC) proliferation. However, the connection between oxidative stress and mitogenic signals remains obscure. In a screen for genes required for ISC proliferation in response to oxidative stress, we identified two regulators of cytosolic Ca2+ levels, transient receptor potential A1 (TRPA1) and ryanodine receptor (RyR). Characterization of TRPA1 and RyR demonstrates that Ca2+ signaling is required for oxidative stress-induced activation of the Ras/MAPK pathway, which in turns drives ISC proliferation. Our findings provide a link between redox regulation and Ca2+ signaling and reveal a novel mechanism by which ISCs detect stress signals.
Collapse
Affiliation(s)
- Chiwei Xu
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Junjie Luo
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, United States.,Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States
| | - Li He
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Craig Montell
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, United States.,Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, United States
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| |
Collapse
|