1
|
DeLoriea J, Millet-Boureima C, Gamberi C. Protocol to build a drug-testing pipeline using large populations of Drosophila melanogaster. STAR Protoc 2023; 4:102747. [PMID: 38103197 PMCID: PMC10751569 DOI: 10.1016/j.xpro.2023.102747] [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: 08/08/2023] [Revised: 10/05/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
As a small animal that recapitulates many fundamental aspects of human disease, Drosophila lends itself to probing the biological activity of molecules and drug candidates. Here, we present a protocol to build a drug-testing pipeline in Drosophila. We describe steps for generating synchronous populations of Bicaudal C mutants by genetic crossing and wild-type fly culturing for controlled compound administration and exemplary phenotypic assays. For complete details on the use and execution of this protocol, please refer to Millet-Boureima et al.,1 Millet-Boureima et al.,2 and Gamberi et al.3.
Collapse
Affiliation(s)
- Jay DeLoriea
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA
| | | | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA.
| |
Collapse
|
2
|
Dowdle ME, Kanzler CR, Harder CRK, Moffet S, Walker MN, Sheets MD. Bicaudal-C Post-transcriptional regulator of cell fates and functions. Front Cell Dev Biol 2022; 10:981696. [PMID: 36158189 PMCID: PMC9491823 DOI: 10.3389/fcell.2022.981696] [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: 06/29/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Bicaudal-C (Bicc1) is an evolutionarily conserved RNA binding protein that functions in a regulatory capacity in a variety of contexts. It was originally identified as a genetic locus in Drosophila that when disrupted resulted in radical changes in early development. In the most extreme phenotypes embryos carrying mutations developed with mirror image duplications of posterior structures and it was this striking phenotype that was responsible for the name Bicaudal. These seminal studies established Bicc1 as an important regulator of Drosophila development. What was not anticipated from the early work, but was revealed subsequently in many different organisms was the broad fundamental impact that Bicc1 proteins have on developmental biology; from regulating cell fates in vertebrate embryos to defects associated with several human disease states. In the following review we present a perspective of Bicc1 focusing primarily on the molecular aspects of its RNA metabolism functions in vertebrate embryos.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael D. Sheets
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
3
|
Dow JAT, Simons M, Romero MF. Drosophila melanogaster: a simple genetic model of kidney structure, function and disease. Nat Rev Nephrol 2022; 18:417-434. [PMID: 35411063 DOI: 10.1038/s41581-022-00561-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/27/2022]
Abstract
Although the genetic basis of many kidney diseases is being rapidly elucidated, their experimental study remains problematic owing to the lack of suitable models. The fruitfly Drosophila melanogaster provides a rapid, ethical and cost-effective model system of the kidney. The unique advantages of D. melanogaster include ease and low cost of maintenance, comprehensive availability of genetic mutants and powerful transgenic technologies, and less onerous regulation, as compared with mammalian systems. Renal and excretory functions in D. melanogaster reside in three main tissues - the transporting renal (Malpighian) tubules, the reabsorptive hindgut and the endocytic nephrocytes. Tubules contain multiple cell types and regions and generate a primary urine by transcellular transport rather than filtration, which is then subjected to selective reabsorption in the hindgut. By contrast, the nephrocytes are specialized for uptake of macromolecules and equipped with a filtering slit diaphragm resembling that of podocytes. Many genes with key roles in the human kidney have D. melanogaster orthologues that are enriched and functionally relevant in fly renal tissues. This similarity has allowed investigations of epithelial transport, kidney stone formation and podocyte and proximal tubule function. Furthermore, a range of unique quantitative phenotypes are available to measure function in both wild type and disease-modelling flies.
Collapse
Affiliation(s)
- Julian A T Dow
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Matias Simons
- INSERM UMR1163, Laboratory of Epithelial Biology and Disease, Imagine Institute, Université de Paris, Hôpital Necker-Enfants Malades, Paris, France
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael F Romero
- Department of Physiology and Biomedical Engineering, Division of Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
4
|
Abstract
RNA-binding proteins (RBPs) are of fundamental importance for post-transcriptional gene regulation and protein synthesis. They are required for pre-mRNA processing and for RNA transport, degradation and translation into protein, and can regulate every step in the life cycle of their RNA targets. In addition, RBP function can be modulated by RNA binding. RBPs also participate in the formation of ribonucleoprotein complexes that build up macromolecular machineries such as the ribosome and spliceosome. Although most research has focused on mRNA-binding proteins, non-coding RNAs are also regulated and sequestered by RBPs. Functional defects and changes in the expression levels of RBPs have been implicated in numerous diseases, including neurological disorders, muscular atrophy and cancers. RBPs also contribute to a wide spectrum of kidney disorders. For example, human antigen R has been reported to have a renoprotective function in acute kidney injury (AKI) but might also contribute to the development of glomerulosclerosis, tubulointerstitial fibrosis and diabetic kidney disease (DKD), loss of bicaudal C is associated with cystic kidney diseases and Y-box binding protein 1 has been implicated in the pathogenesis of AKI, DKD and glomerular disorders. Increasing data suggest that the modulation of RBPs and their interactions with RNA targets could be promising therapeutic strategies for kidney diseases.
Collapse
|
5
|
Zhang P, Azad P, Engelhart DC, Haddad GG, Nigam SK. SLC22 Transporters in the Fly Renal System Regulate Response to Oxidative Stress In Vivo. Int J Mol Sci 2021; 22:13407. [PMID: 34948211 PMCID: PMC8706193 DOI: 10.3390/ijms222413407] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Several SLC22 transporters in the human kidney and other tissues are thought to regulate endogenous small antioxidant molecules such as uric acid, ergothioneine, carnitine, and carnitine derivatives. These transporters include those from the organic anion transporter (OAT), OCTN/OCTN-related, and organic cation transporter (OCT) subgroups. In mammals, it has been difficult to show a clear in vivo role for these transporters during oxidative stress. Ubiquitous knockdowns of related Drosophila SLC22s-including transporters homologous to those previously identified by us in mammals such as the "Fly-Like Putative Transporters" FLIPT1 (SLC22A15) and FLIPT2 (SLC22A16)-have shown modest protection against oxidative stress. However, these fly transporters tend to be broadly expressed, and it is unclear if there is an organ in which their expression is critical. Using two tissue-selective knockdown strategies, we were able to demonstrate much greater and longer protection from oxidative stress compared to previous whole fly knockdowns as well as both parent and WT strains (CG6126: p < 0.001, CG4630: p < 0.01, CG16727: p < 0.0001 and CG6006: p < 0.01). Expression in the Malpighian tubule and likely other tissues as well (e.g., gut, fat body, nervous system) appear critical for managing oxidative stress. These four Drosophila SLC22 genes are similar to human SLC22 transporters (CG6126: SLC22A16, CG16727: SLC22A7, CG4630: SLC22A3, and CG6006: SLC22A1, SLC22A2, SLC22A3, SLC22A6, SLC22A7, SLC22A8, SLC22A11, SLC22A12 (URAT1), SLC22A13, SLC22A14)-many of which are highly expressed in the kidney. Consistent with the Remote Sensing and Signaling Theory, this indicates an important in vivo role in the oxidative stress response for multiple SLC22 transporters within the fly renal system, perhaps through interaction with SLC22 counterparts in non-renal tissues. We also note that many of the human relatives are well-known drug transporters. Our work not only indicates the importance of SLC22 transporters in the fly renal system but also sets the stage for in vivo studies by examining their role in mammalian oxidative stress and organ crosstalk.
Collapse
Affiliation(s)
- Patrick Zhang
- Department of Biology, University of California San Diego, La Jolla, CA 92093, USA; (P.Z.); (D.C.E.)
| | - Priti Azad
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; (P.A.); (G.G.H.)
| | - Darcy C. Engelhart
- Department of Biology, University of California San Diego, La Jolla, CA 92093, USA; (P.Z.); (D.C.E.)
| | - Gabriel G. Haddad
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; (P.A.); (G.G.H.)
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
- Rady Children’s Hospital, San Diego, CA 92123, USA
| | - Sanjay K. Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; (P.A.); (G.G.H.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Han Q, Jiang J, Yuan Y, Tang B, Zhang J. Bicaudal-C protein, a potential antidepressant target. Neuroreport 2021; 32:1293-1298. [PMID: 34554934 DOI: 10.1097/wnr.0000000000001729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Bicaudal-C protein is a highly conserved RNA binding protein, which contains K homology domains and sterile alpha motif domain. Genome-wide association study identified that Bicaudal-C protein was associated with depression. The expression of Bicaudal-C increased in depression patients, also increased expression of Bicaudal-C induces the behavior of depression. The decrease of synaptic plasticity plays a part in depression. Bicaudal-C protein reduces the synaptic plasticity of neurons via TrkB/mTOR/AMPA/pGluA1 pathways, Wnt pathway, or influencing some proteins related to synaptic plasticity. The decreased expression of Bicaudal-C plays an important role in the action of several antidepressants, such as ketamine, biperiden, and scopolamine. Therefore, Bicaudal-C protein may be a potential antidepressant target. Clarifying the relationship between Bicaudal-C protein and depression may help us to find new antidepressants. This review focuses on the research advances of the relationship between Bicaudal-C protein and depression.
Collapse
Affiliation(s)
- Qinghua Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | | | | | | | | |
Collapse
|
7
|
Empowering Melatonin Therapeutics with Drosophila Models. Diseases 2021; 9:diseases9040067. [PMID: 34698120 PMCID: PMC8544433 DOI: 10.3390/diseases9040067] [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: 08/01/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Melatonin functions as a central regulator of cell and organismal function as well as a neurohormone involved in several processes, e.g., the regulation of the circadian rhythm, sleep, aging, oxidative response, and more. As such, it holds immense pharmacological potential. Receptor-mediated melatonin function mainly occurs through MT1 and MT2, conserved amongst mammals. Other melatonin-binding proteins exist. Non-receptor-mediated activities involve regulating the mitochondrial function and antioxidant cascade, which are frequently affected by normal aging as well as disease. Several pathologies display diseased or dysfunctional mitochondria, suggesting melatonin may be used therapeutically. Drosophila models have extensively been employed to study disease pathogenesis and discover new drugs. Here, we review the multiple functions of melatonin through the lens of functional conservation and model organism research to empower potential melatonin therapeutics to treat neurodegenerative and renal diseases.
Collapse
|
8
|
Nita A, Abraham SP, Krejci P, Bosakova M. Oncogenic FGFR Fusions Produce Centrosome and Cilia Defects by Ectopic Signaling. Cells 2021; 10:1445. [PMID: 34207779 PMCID: PMC8227969 DOI: 10.3390/cells10061445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
A single primary cilium projects from most vertebrate cells to guide cell fate decisions. A growing list of signaling molecules is found to function through cilia and control ciliogenesis, including the fibroblast growth factor receptors (FGFR). Aberrant FGFR activity produces abnormal cilia with deregulated signaling, which contributes to pathogenesis of the FGFR-mediated genetic disorders. FGFR lesions are also found in cancer, raising a possibility of cilia involvement in the neoplastic transformation and tumor progression. Here, we focus on FGFR gene fusions, and discuss the possible mechanisms by which they function as oncogenic drivers. We show that a substantial portion of the FGFR fusion partners are proteins associated with the centrosome cycle, including organization of the mitotic spindle and ciliogenesis. The functions of centrosome proteins are often lost with the gene fusion, leading to haploinsufficiency that induces cilia loss and deregulated cell division. We speculate that this complements the ectopic FGFR activity and drives the FGFR fusion cancers.
Collapse
Affiliation(s)
- Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Sara P. Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| |
Collapse
|
9
|
Millet-Boureima C, He S, Le TBU, Gamberi C. Modeling Neoplastic Growth in Renal Cell Carcinoma and Polycystic Kidney Disease. Int J Mol Sci 2021; 22:3918. [PMID: 33920158 PMCID: PMC8070407 DOI: 10.3390/ijms22083918] [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: 03/01/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) and autosomal dominant polycystic kidney disease (ADPKD) share several characteristics, including neoplastic cell growth, kidney cysts, and limited therapeutics. As well, both exhibit impaired vasculature and compensatory VEGF activation of angiogenesis. The PI3K/AKT/mTOR and Ras/Raf/ERK pathways play important roles in regulating cystic and tumor cell proliferation and growth. Both RCC and ADPKD result in hypoxia, where HIF-α signaling is activated in response to oxygen deprivation. Primary cilia and altered cell metabolism may play a role in disease progression. Non-coding RNAs may regulate RCC carcinogenesis and ADPKD through their varied effects. Drosophila exhibits remarkable conservation of the pathways involved in RCC and ADPKD. Here, we review the progress towards understanding disease mechanisms, partially overlapping cellular and molecular dysfunctions in RCC and ADPKD and reflect on the potential for the agile Drosophila genetic model to accelerate discovery science, address unresolved mechanistic aspects of these diseases, and perform rapid pharmacological screens.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Thi Bich Uyen Le
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore 119228, Singapore
| | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29528-6054, USA
| |
Collapse
|
10
|
Pascual A, Vilardo ES, Taibo C, Sabio Y García J, Pomar RR. Bicaudal C is required for the function of the follicular epithelium during oogenesis in Rhodnius prolixus. Dev Genes Evol 2021; 231:33-45. [PMID: 33704576 DOI: 10.1007/s00427-021-00673-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/09/2021] [Indexed: 11/25/2022]
Abstract
The morphology and physiology of the oogenesis have been well studied in the vector of Chagas disease Rhodnius prolixus. However, the molecular interactions that regulate the process of egg formation, key for the reproductive cycle of the vector, is still largely unknown. In order to understand the molecular and cellular basis of the oogenesis, we examined the function of the gene Bicaudal C (BicC) during oogenesis and early development of R. prolixus. We show that R. prolixus BicC (Rp-BicC) gene is expressed in the germarium, with cytoplasmic distribution, as well as in the follicular epithelium of the developing oocytes. RNAi silencing of Rp-BicC resulted in sterile females that lay few, small, non-viable eggs. The ovaries are reduced in size and show a disarray of the follicular epithelium. This indicates that Rp-BicC has a central role in the regulation of oogenesis. Although the follicular cells are able to form the chorion, the uptake of vitelline by the oocytes is compromised. We show evidence that the polarity of the follicular epithelium and the endocytic pathway, which are crucial for the proper yolk deposition, are affected. This study provides insights into the molecular mechanisms underlying oocyte development and show that Rp-BicC is important for de developmental of the egg and, therefore, a key player in the reproduction of this insect.
Collapse
Affiliation(s)
- Agustina Pascual
- Centro de Bioinvestigaciones (UNNOBA-CICBA), CITNOBA (UNNOBA-CONICET), 2700, Pergamino, Buenos Aires, Argentina.
| | - Emiliano S Vilardo
- Centro Regional de Estudios Genómicos (Facultad de Ciencias Exactas, CREG-UNLP), 1900, La Plata, Buenos Aires, Argentina
| | - Catalina Taibo
- Laboratorio de Microscopia Integral (LIM), (CICVyA, INTA), 1686, Hurlingham, Buenos Aires, Argentina
| | - Julia Sabio Y García
- Laboratorio de Microscopia Integral (LIM), (CICVyA, INTA), 1686, Hurlingham, Buenos Aires, Argentina
| | - Rolando Rivera Pomar
- Centro de Bioinvestigaciones (UNNOBA-CICBA), CITNOBA (UNNOBA-CONICET), 2700, Pergamino, Buenos Aires, Argentina.
- Centro Regional de Estudios Genómicos (Facultad de Ciencias Exactas, CREG-UNLP), 1900, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Cyst Reduction by Melatonin in a Novel Drosophila Model of Polycystic Kidney Disease. Molecules 2020; 25:molecules25225477. [PMID: 33238462 PMCID: PMC7700119 DOI: 10.3390/molecules25225477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) causes progressive cystic degeneration of the renal tubules, the nephrons, eventually severely compromising kidney function. ADPKD is incurable, with half of the patients eventually needing renal replacement. Treatments for ADPKD patients are limited and new effective therapeutics are needed. Melatonin, a central metabolic regulator conserved across all life kingdoms, exhibits oncostatic and oncoprotective activity and no detected toxicity. Here, we used the Bicaudal C (BicC) Drosophila model of polycystic kidney disease to test the cyst-reducing potential of melatonin. Significant cyst reduction was found in the renal (Malpighian) tubules upon melatonin administration and suggest mechanistic sophistication. Similar to vertebrate PKD, the BicC fly PKD model responds to the antiproliferative drugs rapamycin and mimics of the second mitochondria-derived activator of caspases (Smac). Melatonin appears to be a new cyst-reducing molecule with attractive properties as a potential candidate for PKD treatment.
Collapse
|
12
|
Zhao R, Peng C, Song C, Zhao Q, Rong J, Wang H, Ding W, Wang F, Xie Y. BICC1 as a novel prognostic biomarker in gastric cancer correlating with immune infiltrates. Int Immunopharmacol 2020; 87:106828. [PMID: 32736193 DOI: 10.1016/j.intimp.2020.106828] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 12/24/2022]
Abstract
AIM BicC family RNA-binding protein 1 (BICC1) codes an RNA-binding protein that regulates gene expression and modulates cell proliferation and apoptosis. We aim at investigating the role of BICC1 in gastric carcinogenesis. METHODS BICC1 mRNA expression in gastric cancer (GC) was examined using the Tumor Immune Estimation Resource (TIMER), The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Correlations between BICC1 expression and clinicopathological parameters were analyzed. The Gene Expression Profiling Interactive Analysis (GEPIA) and Kaplan-Meier plotter databases were used to examine the clinical prognostic significance of BICC1 in GC. Signaling pathways related to BICC1 expression were identified by gene set enrichment analysis (GSEA). TIMER and CIBERSORT were used to analyze the correlations among BICC1, BICC1-coexpressed genes and tumor-infiltrating immune cells. RESULTS BICC1 was highly expressed in GC and significantly correlated with grade (P = 0.002), TNM stage (P = 0.033), invasion depth (P = 0.001) and vital status (P = 0.009) of GC patients. High BICC1 expression correlated with poor overall survival. The GSEA results showed that cell adhesion-, tumor- and immune- related pathways were significantly enriched in samples with high BICC1 expression. BICC1 and its coexpressed genes were positively related to tumor-infiltrating immune cells and were strongly correlated with tumor-infiltrating macrophages (all r ≥ 0.582, P < 0.0001). The CIBERSORT database revealed that BICC1 correlated with M2 macrophages (P < 0.0001), regulatory T cells (P < 0.0001), resting mast cells (P < 0.0001), activated memory CD4+ T cells (P = 0.002), resting NK cells (P = 0.002), activated dendritic cells (P = 0.002), and follicular helper T cells (P = 0.016). The results from TIMER database confirmed that BICC1 is closely associated with the markers of M2 macrophages and tumor-associated macrophages (all r ≥ 0.5, P < 0.0001). CONCLUSION BICC1 may be a potential prognostic biomarker in GC and correlates with immune infiltrates.
Collapse
Affiliation(s)
- Rulin Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China
| | - Chao Peng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China
| | - Conghua Song
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China
| | - Qiaoyun Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China
| | - Jianfang Rong
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China
| | - Huan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China
| | - Wenjie Ding
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1, Min de Street, Nanchang 330000, Jiangxi, China
| | - Fangfei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330000, Jiangxi, China.
| |
Collapse
|
13
|
An Overview of In Vivo and In Vitro Models for Autosomal Dominant Polycystic Kidney Disease: A Journey from 3D-Cysts to Mini-Pigs. Int J Mol Sci 2020; 21:ijms21124537. [PMID: 32630605 PMCID: PMC7352572 DOI: 10.3390/ijms21124537] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inheritable cause of end stage renal disease and, as of today, only a single moderately effective treatment is available for patients. Even though ADPKD research has made huge progress over the last decades, the precise disease mechanisms remain elusive. However, a wide variety of cellular and animal models have been developed to decipher the pathophysiological mechanisms and related pathways underlying the disease. As none of these models perfectly recapitulates the complexity of the human disease, the aim of this review is to give an overview of the main tools currently available to ADPKD researchers, as well as their main advantages and limitations.
Collapse
|
14
|
Millet-Boureima C, Selber-Hnatiw S, Gamberi C. Drug discovery and chemical probing in Drosophila. Genome 2020; 64:147-159. [PMID: 32551911 DOI: 10.1139/gen-2020-0037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Flies are increasingly utilized in drug discovery and chemical probing in vivo, which are novel technologies complementary to genetic probing in fundamental biological studies. Excellent genetic conservation, small size, short generation time, and over one hundred years of genetics make Drosophila an attractive model for rapid assay readout and use of analytical amounts of compound, enabling the experimental iterations needed in early drug development at a fraction of time and costs. Here, we describe an effective drug-testing pipeline using adult flies that can be easily implemented to study several disease models and different genotypes to discover novel molecular insight, probes, quality lead compounds, and develop novel prototype drugs.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Susannah Selber-Hnatiw
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
15
|
Parrot C, Kurbegovic A, Yao G, Couillard M, Côté O, Trudel M. c-Myc is a regulator of the PKD1 gene and PC1-induced pathogenesis. Hum Mol Genet 2020; 28:751-763. [PMID: 30388220 DOI: 10.1093/hmg/ddy379] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/28/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is among the most common monogenic disorders mainly associated with PKD1/PC1 mutations. We show herein that renal regulation in Pc1 dosage-reduced and -increased mouse models converge toward stimulation of c-Myc expression along with β-catenin, delineating c-Myc as a key Pkd1 node in cystogenesis. Enhanced renal c-Myc-induced ADPKD in SBM transgenic mice lead conversely to striking upregulation of Pkd1/Pc1 expression and β-catenin activation, lending credence for reciprocal crosstalk between c-Myc and Pc1. In adult SBM kidneys, c-Myc is strongly enriched on Pkd1 promoter with RNA pol II, consistent with Pkd1 upregulation during cystogenesis. Similar c-Myc direct binding at birth uncovers an equivalent role on Pkd1 regulation during renal developmental program. Concurrent with enriched c-Myc binding, recruitment of active chromatin modifying co-factors by c-Myc at the Pkd1 regulatory region probably opens chromatin to stimulate transcription. A similar transcriptional activation by c-Myc is also likely operant on endogenous human PKD1 gene from our transactivation analysis in response to human c-MYC upregulation. Genetic ablation of c-Myc in Pc1-reduced and -increased mouse models significantly attenuates cyst growth, proliferation and PKD progression. Our study determined a dual role for c-Myc, as a major contributor in Pc1-induced cystogenesis and in a feed-forward regulatory Pkd1-c-Myc loop mechanism that may also prevail in human ADPKD.
Collapse
Affiliation(s)
- Camila Parrot
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Almira Kurbegovic
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Guanhan Yao
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Martin Couillard
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Olivier Côté
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Marie Trudel
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
16
|
Kurbegovic A, Trudel M. The master regulators Myc and p53 cellular signaling and functions in polycystic kidney disease. Cell Signal 2020; 71:109594. [PMID: 32145315 DOI: 10.1016/j.cellsig.2020.109594] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 01/08/2023]
Abstract
The transcription factors Myc and p53 associated with oncogenesis play determinant roles in a human genetic disorder, autosomal dominant polycystic kidney disease (ADPKD), that was coined early in ADPKD etiology a «neoplasia in disguise ». These factors are interdependent master cell regulators of major biological processes including proliferation, apoptosis, cell growth, metabolism, inflammation, fibrosis and differentiation that are all modulated in ADPKD. Myc and p53 proteins evolved to respond and carry out overlapping functions via opposing mechanisms of action. Studies in human ADPKD kidneys, caused by mutations in the PKD1 or PKD2 genes, reveal reduced p53 expression and high expression of Myc in the cystic tubular epithelium. Myc and p53 via direct interaction act respectively, as transcriptional activator and repressor of PKD1 gene expression, consistent with increased renal PKD1 levels in ADPKD. Mouse models generated by Pkd1 and Pkd2 gene dosage dysregulation reproduce renal cystogenesis with activation of Myc expression and numerous signaling pathways, strikingly similar to those determined in human ADPKD. In fact, upregulation of renal Myc expression is also detected in virtually all non-orthologous animal models of PKD. A definitive causal connection of Myc with cystogenesis was established by renal overexpression of Myc in transgenic mice that phenocopies human ADPKD. The network of activated signaling pathways in human and mouse cystogenesis individually or in combination can target Myc as a central node of PKD pathogenesis. One or many of the multiple functions of Myc upon activation can play a role in every phases of ADPKD development and lend credence to the notion of "Myc addiction" for cystogenesis. We propose that the residual p53 levels are conducive to an ADPKD biological program without cancerogenesis while a "p53 dependent annihilation" mechanism would be permissive to oncogenesis. Of major importance, Myc ablation in orthologous mouse models or direct inhibition in non-orthologous mouse model significantly delays cystogenesis consistent with pharmacologic or genetic inhibition of Myc upstream regulator or downstream targets in the mouse. Together, these studies on PKD proteins upon dysregulation not only converged on Myc as a focal point but also attribute to Myc upregulation a causal and « driver » role in pathogenesis. This review will present and discuss our current knowledge on Myc and p53, focused on PKD mouse models and ADPKD.
Collapse
Affiliation(s)
- Almira Kurbegovic
- Institut de Recherches Cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Marie Trudel
- Institut de Recherches Cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
17
|
Cohen E, Sawyer JK, Peterson NG, Dow JAT, Fox DT. Physiology, Development, and Disease Modeling in the Drosophila Excretory System. Genetics 2020; 214:235-264. [PMID: 32029579 PMCID: PMC7017010 DOI: 10.1534/genetics.119.302289] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
The insect excretory system contains two organ systems acting in concert: the Malpighian tubules and the hindgut perform essential roles in excretion and ionic and osmotic homeostasis. For over 350 years, these two organs have fascinated biologists as a model of organ structure and function. As part of a recent surge in interest, research on the Malpighian tubules and hindgut of Drosophila have uncovered important paradigms of organ physiology and development. Further, many human disease processes can be modeled in these organs. Here, focusing on discoveries in the past 10 years, we provide an overview of the anatomy and physiology of the Drosophila excretory system. We describe the major developmental events that build these organs during embryogenesis, remodel them during metamorphosis, and repair them following injury. Finally, we highlight the use of the Malpighian tubules and hindgut as accessible models of human disease biology. The Malpighian tubule is a particularly excellent model to study rapid fluid transport, neuroendocrine control of renal function, and modeling of numerous human renal conditions such as kidney stones, while the hindgut provides an outstanding model for processes such as the role of cell chirality in development, nonstem cell-based injury repair, cancer-promoting processes, and communication between the intestine and nervous system.
Collapse
Affiliation(s)
| | - Jessica K Sawyer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, and
| | | | - Julian A T Dow
- Institute of Molecular, Cell, and Systems Biology, University of Glasgow, G12 8QQ, United Kingdom
| | - Donald T Fox
- Department of Cell Biology and
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, and
| |
Collapse
|
18
|
Rothé B, Gagnieux C, Leal-Esteban LC, Constam DB. Role of the RNA-binding protein Bicaudal-C1 and interacting factors in cystic kidney diseases. Cell Signal 2019; 68:109499. [PMID: 31838063 DOI: 10.1016/j.cellsig.2019.109499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/03/2023]
Abstract
Polycystic kidneys frequently associate with mutations in individual components of cilia, basal bodies or centriolar satellites that perturb complex protein networks. In this review, we focus on the RNA-binding protein Bicaudal-C1 (BICC1) which was found mutated in renal cystic dysplasia, and on its interactions with the ankyrin repeat and sterile α motif (SAM)-containing proteins ANKS3 and ANKS6 and associated kinases and their partially overlapping ciliopathy phenotypes. After reviewing BICC1 homologs in model organisms and their functions in mRNA and cell metabolism during development and in renal tubules, we discuss recent insights from cell-based assays and from structure analysis of the SAM domains, and how SAM domain oligomerization might influence multivalent higher order complexes that are implicated in ciliary signal transduction.
Collapse
Affiliation(s)
- Benjamin Rothé
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland
| | - Céline Gagnieux
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland
| | - Lucia Carolina Leal-Esteban
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland; Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Daniel B Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Station 19, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
19
|
Millet-Boureima C, Chingle R, Lubell WD, Gamberi C. Cyst Reduction in a Polycystic Kidney Disease Drosophila Model Using Smac Mimics. Biomedicines 2019; 7:biomedicines7040082. [PMID: 31635379 PMCID: PMC6966561 DOI: 10.3390/biomedicines7040082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited malady affecting 12.5 million people worldwide. Therapeutic options to treat PKD are limited, due in part to lack of precise knowledge of underlying pathological mechanisms. Mimics of the second mitochondria-derived activator of caspases (Smac) have exhibited activity as antineoplastic agents and reported recently to ameliorate cysts in a murine ADPKD model, possibly by differentially targeting cystic cells and sparing the surrounding tissue. A first-in-kind Drosophila PKD model has now been employed to probe further the activity of novel Smac mimics. Substantial reduction of cystic defects was observed in the Malpighian (renal) tubules of treated flies, underscoring mechanistic conservation of the cystic pathways and potential for efficient testing of drug prototypes in this PKD model. Moreover, the observed differential rescue of the anterior and posterior tubules overall, and within their physiologically diverse intermediate and terminal regions implied a nuanced response in distinct tubular regions contingent upon the structure of the Smac mimic. Knowledge gained from studying Smac mimics reveals the capacity for the Drosophila model to precisely probe PKD pharmacology highlighting the value for such critical evaluation of factors implicated in renal function and pathology.
Collapse
Affiliation(s)
| | - Ramesh Chingle
- Département de Chimie, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| | - William D Lubell
- Département de Chimie, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.
| |
Collapse
|
20
|
Foo JN, Xia Y. Polycystic kidney disease: new knowledge and future promises. Curr Opin Genet Dev 2019; 56:69-75. [PMID: 31476629 DOI: 10.1016/j.gde.2019.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/27/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023]
Abstract
Polycystic kidney disease (PKD) is one of the most common genetic kidney diseases, characterized by the formation of fluid-filled renal cysts, which eventually lead to end-stage renal disease. Despite several decades of investigation, explicit molecular and cellular mechanisms underpinning renal cyst formation have been unresolved until recently, severely hampering the development of effective therapeutic approaches. Currently, most PKD therapies have been developed for limiting disease complications, such as hypertension. Although Tolvaptan has been approved for treating PKD in few countries, the associated hepatic toxicity remains a major concern. In this Review, we will discuss recent advances in PKD research, covering aspects ranging from newly identified genetic/epigenetic causes, increment in mechanistic interpretation, novel therapeutic targets, to the promises offered by emerging stem cell technologies.
Collapse
Affiliation(s)
- Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, 308232, Singapore; Human Genetics, Genome Institute of Singapore, A(⁎)STAR, 138672, Singapore.
| | - Yun Xia
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, 308232, Singapore.
| |
Collapse
|
21
|
Yang E, Gamberi C, Chaurand P. Mapping the fly Malpighian tubule lipidome by imaging mass spectrometry. JOURNAL OF MASS SPECTROMETRY : JMS 2019; 54:557-566. [PMID: 31038251 DOI: 10.1002/jms.4366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 05/20/2023]
Abstract
Matrix-assisted laser/desorption ionization imaging mass spectrometry (MALDI IMS) is an analytical technique for understanding the spatial distribution of biomolecules across a sample surface. Originally employed for mammalian tissues, this technology has been adapted to study specimens as diverse as microbes and cell cultures, food such as strawberries, and invertebrates including the vinegar fly Drosophila melanogaster. As an ideal model organism, Drosophila has brought greater understanding about conserved biological processes, organism development, and diseased states and even informed management practices of agriculturally and environmentally important species. Drosophila displays anatomically separated renal (Malpighian) tubules that are the physiological equivalent to the vertebrate nephron. Insect Malpighian tubules are also responsible for pesticide detoxification. In this article, we first describe an effective workflow and sample preparation method to study the phospholipid distribution of the Malpighian tubules that initially involves the manual microdissection of the tubules in saline buffer followed by a series of washes to remove excess salt and enhances the phospholipid signals prior to matrix deposition and IMS at 25-μm spatial resolution. We also established a complementary methodology for lipid IMS analysis of whole-body fly sections using a dual-polarity data acquisition approach at the same spatial resolution after matrix deposition by sublimation. Both procedures yield rich signal profiles from the major phospholipid classes. The reproducibility and high-quality results offered by these methodologies enable cohort studies of Drosophila through MALDI IMS.
Collapse
Affiliation(s)
- Ethan Yang
- Department of Chemistry, University of Montreal, Pavillon Roger-Gaudry, 2900, boul. Édouard-Montpetit, Montreal, QC, Canada, H3C 3J7
| | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC, Canada, H4B 1R6
| | - Pierre Chaurand
- Department of Chemistry, University of Montreal, Pavillon Roger-Gaudry, 2900, boul. Édouard-Montpetit, Montreal, QC, Canada, H3C 3J7
| |
Collapse
|
22
|
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: 30] [Impact Index Per Article: 5.0] [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
|
23
|
Wei D, Zhang YX, Liu YW, Li WJ, Chen ZX, Smagghe G, Wang JJ. Gene expression profiling of ovary identified eggshell proteins regulated by 20-hydroxyecdysone in Bactrocera dorsalis. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2019; 30:206-216. [PMID: 30909163 DOI: 10.1016/j.cbd.2019.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 02/03/2023]
Abstract
The oriental fruit fly, Bactrocera dorsalis, is one of the most destructive pests worldwide. The frequent use of chemical insecticides has led B. dorsalis to develop resistance to many insecticides in recent decades. New high-throughput-sequenced transcriptomes, as well as genomes, have revealed a large number of reference genes for functional target identification. Here, we performed digital gene expression profiling of ovary and testis of B. dorsalis adults. Various genes were identified to be highly expressed in B. dorsalis ovary. The genes encoding components of eggshell, vitelline membrane proteins (Vmps) and chorion-related proteins, were identified to be tissue-specifically expressed in ovary. Five cytochrome P450 genes were also identified to be highly expressed in ovary. Three of them were ecdysone synthesis pathway genes indicating the ovary as a potential synthesis site of female. The up-regulated expression of Vmps by exogenous 20-hydroxyecdysone implied the hormonal regulation of eggshell formation during ovarian development. Many other genes with potential functions in ovarian development were also identified, including vitellogenin receptor, insulin receptor, NASP protein, and odorant binding protein. These findings should promote our understanding of the regulation of vitellogenesis and eggshell formation and enable exploration of potentially novel pest control targets.
Collapse
Affiliation(s)
- Dong Wei
- Key Laboratory of Entomology and Pest Control Engineering of Chongqing, College of Plant Protection, Southwest University, Chongqing 400715, China; International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China; Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Ying-Xin Zhang
- Key Laboratory of Entomology and Pest Control Engineering of Chongqing, College of Plant Protection, Southwest University, Chongqing 400715, China; International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Yu-Wei Liu
- Key Laboratory of Entomology and Pest Control Engineering of Chongqing, College of Plant Protection, Southwest University, Chongqing 400715, China; International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Wei-Jun Li
- Key Laboratory of Entomology and Pest Control Engineering of Chongqing, College of Plant Protection, Southwest University, Chongqing 400715, China; International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Zhi-Xian Chen
- Key Laboratory of Entomology and Pest Control Engineering of Chongqing, College of Plant Protection, Southwest University, Chongqing 400715, China; International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Guy Smagghe
- Key Laboratory of Entomology and Pest Control Engineering of Chongqing, College of Plant Protection, Southwest University, Chongqing 400715, China; International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China; Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Jin-Jun Wang
- Key Laboratory of Entomology and Pest Control Engineering of Chongqing, College of Plant Protection, Southwest University, Chongqing 400715, China; International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
24
|
Leal-Esteban LC, Rothé B, Fortier S, Isenschmid M, Constam DB. Role of Bicaudal C1 in renal gluconeogenesis and its novel interaction with the CTLH complex. PLoS Genet 2018; 14:e1007487. [PMID: 29995892 PMCID: PMC6056059 DOI: 10.1371/journal.pgen.1007487] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/23/2018] [Accepted: 06/13/2018] [Indexed: 01/06/2023] Open
Abstract
Altered glucose and lipid metabolism fuel cystic growth in polycystic kidneys, but the cause of these perturbations is unclear. Renal cysts also associate with mutations in Bicaudal C1 (Bicc1) or in its self-polymerizing sterile alpha motif (SAM). Here, we found that Bicc1 maintains normoglycemia and the expression of the gluconeogenic enzymes FBP1 and PEPCK in kidneys. A proteomic screen revealed that Bicc1 interacts with the C-Terminal to Lis-Homology domain (CTLH) complex. Since the orthologous Gid complex in S. cerevisae targets FBP1 and PEPCK for degradation, we mapped the topology among CTLH subunits and found that SAM-mediated binding controls Bicc1 protein levels, whereas Bicc1 inhibited the accumulation of several CTLH subunits. Under the conditions analyzed, Bicc1 increased FBP1 protein levels independently of the CTLH complex. Besides linking Bicc1 to cell metabolism, our findings reveal new layers of complexity in the regulation of renal gluconeogenesis compared to lower eukaryotes. Polycystic kidney diseases (PKD) are incurable inherited chronic disorders marked by fluid-filled cysts that frequently cause renal failure. A glycolytic metabolism reminiscent of cancerous cells accelerates cystic growth, but the mechanism underlying such metabolic re-wiring is poorly understood. PKD-like cystic kidneys also develop in mice that lack the RNA-binding protein Bicaudal-C (Bicc1), and mutations in a single copy of human BICC1 associate with renal cystic dysplasia. Here, we report that Bicc1 regulates renal gluconeogenesis. A screen for interacting factors revealed that Bicc1 binds the C-Terminal to Lis-Homology domain (CTLH) complex, which in lower eukaryotes mediates degradation of gluconeogenic enzymes. By contrast, Bicc1 and the mammalian CTLH complex regulated each other, and Bicc1 stimulated the accumulation of the rate-limiting gluconeogenic enzyme even in cells depleted of CTLH subunits. Our finding that Bicc1 is required for normoglycemia implies that renal gluconeogenesis may be important to inhibit cyst formation.
Collapse
Affiliation(s)
- Lucia Carolina Leal-Esteban
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Benjamin Rothé
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Simon Fortier
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Manuela Isenschmid
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
| | - Daniel B. Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
25
|
Modeling Renal Disease "On the Fly". BIOMED RESEARCH INTERNATIONAL 2018; 2018:5697436. [PMID: 29955604 PMCID: PMC6000847 DOI: 10.1155/2018/5697436] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/17/2018] [Indexed: 12/22/2022]
Abstract
Detoxification is a fundamental function for all living organisms that need to excrete catabolites and toxins to maintain homeostasis. Kidneys are major organs of detoxification that maintain water and electrolyte balance to preserve physiological functions of vertebrates. In insects, the renal function is carried out by Malpighian tubules and nephrocytes. Due to differences in their circulation, the renal systems of mammalians and insects differ in their functional modalities, yet carry out similar biochemical and physiological functions and share extensive genetic and molecular similarities. Evolutionary conservation can be leveraged to model specific aspects of the complex mammalian kidney function in the genetic powerhouse Drosophila melanogaster to study how genes interact in diseased states. Here, we compare the human and Drosophila renal systems and present selected fly disease models.
Collapse
|
26
|
Billot K, Coquil C, Villiers B, Josselin-Foll B, Desban N, Delehouzé C, Oumata N, Le Meur Y, Boletta A, Weimbs T, Grosch M, Witzgall R, Saunier S, Fischer E, Pontoglio M, Fautrel A, Mrug M, Wallace D, Tran PV, Trudel M, Bukanov N, Ibraghimov-Beskrovnaya O, Meijer L. Casein kinase 1ε and 1α as novel players in polycystic kidney disease and mechanistic targets for (R)-roscovitine and (S)-CR8. Am J Physiol Renal Physiol 2018. [PMID: 29537311 DOI: 10.1152/ajprenal.00489.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Following the discovery of (R)-roscovitine's beneficial effects in three polycystic kidney disease (PKD) mouse models, cyclin-dependent kinases (CDKs) inhibitors have been investigated as potential treatments. We have used various affinity chromatography approaches to identify the molecular targets of roscovitine and its more potent analog (S)-CR8 in human and murine polycystic kidneys. These methods revealed casein kinases 1 (CK1) as additional targets of the two drugs. CK1ε expression at the mRNA and protein levels is enhanced in polycystic kidneys of 11 different PKD mouse models as well as in human polycystic kidneys. A shift in the pattern of CK1α isoforms is observed in all PKD mouse models. Furthermore, the catalytic activities of both CK1ε and CK1α are increased in mouse polycystic kidneys. Inhibition of CK1ε and CK1α may thus contribute to the long-lasting attenuating effects of roscovitine and (S)-CR8 on cyst development. CDKs and CK1s may constitute a dual therapeutic target to develop kinase inhibitory PKD drug candidates.
Collapse
Affiliation(s)
- Katy Billot
- ManRos Therapeutics, Centre de Perharidy , Roscoff , France
| | | | | | - Béatrice Josselin-Foll
- CNRS "Protein Phosphorylation and Human Disease Group, Station Biologique, Roscoff Cedex, Bretagne , France
| | - Nathalie Desban
- CNRS "Protein Phosphorylation and Human Disease Group, Station Biologique, Roscoff Cedex, Bretagne , France
| | - Claire Delehouzé
- CNRS "Protein Phosphorylation and Human Disease Group, Station Biologique, Roscoff Cedex, Bretagne , France
| | - Nassima Oumata
- ManRos Therapeutics, Centre de Perharidy , Roscoff , France
| | - Yannick Le Meur
- Service de Néphrologie, Centre Hospitalier Universitaire La Cavale Blanche, Rue Tanguy Prigent, Brest Cedex, France
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, DIBIT San Raffaele Scientific Institute , Milan , Italy
| | - Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California Santa Barbara , Santa Barbara, California
| | - Melanie Grosch
- University of Regensburg, Institute for Molecular and Cellular Anatomy, Universitätsstr 31, Regensburg , Germany
| | - Ralph Witzgall
- University of Regensburg, Institute for Molecular and Cellular Anatomy, Universitätsstr 31, Regensburg , Germany
| | | | - Evelyne Fischer
- "Expression Génique, Développement et Maladies", Equipe 26/INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Institut Cochin, Département Génétique & Développement, Paris , France
| | - Marco Pontoglio
- "Expression Génique, Développement et Maladies", Equipe 26/INSERM U1016/CNRS UMR 8104/Université Paris-Descartes, Institut Cochin, Département Génétique & Développement, Paris , France
| | - Alain Fautrel
- Université de Rennes 1, H2P2 Histopathology Core Facility, Rennes Cedex, France
| | - Michal Mrug
- Division of Nephrology, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Veterans Affairs Medical Center , Birmingham, Alabama
| | - Darren Wallace
- University of Kansas Medical Center, The Jared Grantham Kidney Institute , Kansas City, Kansas
| | - Pamela V Tran
- University of Kansas Medical Center, The Jared Grantham Kidney Institute , Kansas City, Kansas.,University of Kansas Medical Center, Department of Anatomy and Cell Biology , Kansas City, Kansas
| | - Marie Trudel
- Institut de Recherches Cliniques de Montréal, Molecular Genetics and Development, Montreal, Quebec , Canada
| | - Nikolay Bukanov
- Sanofi Genzyme, Rare Renal and Bone Diseases, Framingham, Massachusetts
| | | | - Laurent Meijer
- ManRos Therapeutics, Centre de Perharidy , Roscoff , France
| |
Collapse
|