1
|
Limonta G, Panti C, Fossi MC, Nardi F, Baini M. Exposure to virgin and marine incubated microparticles of biodegradable and conventional polymers modulates the hepatopancreas transcriptome of Mytilus galloprovincialis. JOURNAL OF HAZARDOUS MATERIALS 2024; 468:133819. [PMID: 38402680 DOI: 10.1016/j.jhazmat.2024.133819] [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: 12/05/2023] [Revised: 02/02/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
Biodegradable polymers have been proposed as an alternative to conventional plastics to mitigate the impact of marine litter, but the research investigating their toxicity is still in its infancy. This study evaluates the potential ecotoxicological effects of both virgin and marine-incubated microparticles (MPs), at environmentally relevant concentration (0.1 mg/l), made of different biodegradable polymers (Polycaprolactone, Mater-Bi, cellulose) and conventional polymers (Polyethylene) on Mytilus galloprovincialis by using transcriptomics. This approach is increasingly being used to assess the effects of pollutants on organisms, obtaining data on numerous biological pathways simultaneously. Whole hepatopancreas de novo transcriptome sequencing was performed, individuating 972 genes differentially expressed across experimental groups compared to the control. Through the comparative transcriptomic profiling emerges that the preponderant effect is attributable to the marine incubation of MPs, especially for incubated polycaprolactone (731 DEGs). Mater-Bi and cellulose alter the smallest number of genes and biological processes in the mussel hepatopancreas. All microparticles, regardless of their polymeric composition, dysregulated innate immunity, and fatty acid metabolism biological processes. These findings highlight the necessity of considering the interactions of MPs with the environmental factors in the marine ecosystem when performing ecotoxicological evaluations. The results obtained contribute to fill current knowledge gaps regarding the potential environmental impacts of biodegradable polymers.
Collapse
Affiliation(s)
- Giacomo Limonta
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Cristina Panti
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy.
| | - Maria Cristina Fossi
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Francesco Nardi
- National Biodiversity Future Center (NBFC), Palermo, Italy; Department of Life Sciences, University of Siena, Via A. Moro, 2, Siena, Italy
| | - Matteo Baini
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy
| |
Collapse
|
2
|
Jin R, Forbes CM, Miller NL, Lafin J, Strand DW, Case T, Cates JM, Liu Q, Ramirez-Solano M, Mohler JL, Matusik RJ. Transcriptomic analysis of benign prostatic hyperplasia identifies critical pathways in prostatic overgrowth and 5-alpha reductase inhibitor resistance. Prostate 2024; 84:441-459. [PMID: 38168866 DOI: 10.1002/pros.24661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/06/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The medical therapy of prostatic symptoms (MTOPS) trial randomized men with symptoms of benign prostatic hyperplasia (BPH) and followed response of treatment with a 5α-reductase inhibitor (5ARI), an alpha-adrenergic receptor antagonist (α-blocker), the combination of 5ARI and α-blocker or no medical therapy (none). Medical therapy reduced risk of clinical progression by 66% but the reasons for nonresponse or loss of therapeutic response in some patients remains unresolved. Our previous work showed that prostatic glucocorticoid levels are increased in 5ARI-treated patients and that glucocorticoids can increased branching of prostate epithelia in vitro. To understand the transcriptomic changes associated with 5ARI treatment, we performed bulk RNA sequencing of BPH and control samples from patients who received 5ARI versus those that did not. Deconvolution analysis was performed to estimate cellular composition. Bulk RNA sequencing was also performed on control versus glucocorticoid-treated prostate epithelia in 3D culture to determine underlying transcriptomic changes associated with branching morphogenesis. METHOD Surgical BPH (S-BPH) tissue was defined as benign prostatic tissue collected from the transition zone (TZ) of patients who failed medical therapy while control tissue termed Incidental BPH (I-BPH) was obtained from the TZ of men undergoing radical prostatectomy for low-volume/grade prostatic adenocarcinoma confined to the peripheral zone. S-BPH patients were divided into four subgroups: men on no medical therapy (none: n = 7), α-blocker alone (n = 10), 5ARI alone (n = 6) or combination therapy (α-blocker and 5ARI: n = 7). Control I-BPH tissue was from men on no medical therapy (none: n = 8) or on α-blocker (n = 6). A human prostatic cell line in 3D culture that buds and branches was used to identify genes involved in early prostatic growth. Snap-frozen prostatic tissue taken at the time of surgery and 3D organoids were used for RNA-seq analysis. Bulk RNAseq data were deconvoluted using CIBERSORTx. Differentially expressed genes (DEG) that were statistically significant among S-BPH, I-BPH, and during budding and branching of organoids were used for pathway analysis. RESULTS Transcriptomic analysis between S-BPH (n = 30) and I-BPH (n = 14) using a twofold cutoff (p < 0.05) identified 377 DEG (termed BPH377) and a cutoff < 0.05 identified 3377 DEG (termed BPH3377). Within the S-BPH, the subgroups none and α-blocker were compared to patients on 5ARI to reveal 361 DEG (termed 5ARI361) that were significantly changed. Deconvolution analysis of bulk RNA seq data with a human prostate single cell data set demonstrated increased levels of mast cells, NK cells, interstitial fibroblasts, and prostate luminal cells in S-BPH versus I-BPH. Glucocorticoid (GC)-induced budding and branching of benign prostatic cells in 3D culture was compared to control organoids to identify early events in prostatic morphogenesis. GC induced 369 DEG (termed GC359) in 3D culture. STRING analysis divided the large datasets into 20-80 genes centered around a hub. In general, biological processes induced in BPH supported growth and differentiation such as chromatin modification and DNA repair, transcription, cytoskeleton, mitochondrial electron transport, ubiquitination, protein folding, and cholesterol synthesis. Identified signaling pathways were pooled to create a list of DEG that fell into seven hubs/clusters. The hub gene centrality was used to name the network including AP-1, interleukin (IL)-6, NOTCH1 and NOTCH3, NEO1, IL-13, and HDAC/KDM. All hubs showed connections to inflammation, chromatin structure, and development. The same approach was applied to 5ARI361 giving multiple networks, but the EGF and sonic hedgehog (SHH) hub was of particular interest as a developmental pathway. The BPH3377, 5ARI363, and GC359 lists were compared and 67 significantly changed DEG were identified. Common genes to the 3D culture included an IL-6 hub that connected to genes identified in BPH hubs that defined AP1, IL-6, NOTCH, NEO1, IL-13, and HDAC/KDM. CONCLUSIONS Reduction analysis of BPH and 3D organoid culture uncovered networks previously identified in prostatic development as being reinitiated in BPH. Identification of these pathways provides insight into the failure of medical therapy for BPH and new therapeutic targets for BPH/LUTS.
Collapse
Affiliation(s)
- Renjie Jin
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Connor M Forbes
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Urology Department, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicole L Miller
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Lafin
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Douglas W Strand
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas Case
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M Cates
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marisol Ramirez-Solano
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Robert J Matusik
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
3
|
Tsai YC, Cheng KH, Jiang SS, Hawse JR, Chuang SE, Chen SL, Huang TS, Ch'ang HJ. Krüppel-like factor 10 modulates stem cell phenotypes of pancreatic adenocarcinoma by transcriptionally regulating notch receptors. J Biomed Sci 2023; 30:39. [PMID: 37308977 DOI: 10.1186/s12929-023-00937-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/05/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PDAC) is well known for its rapid distant metastasis and local destructive behavior. Loss of Krüppel-like factor 10 (KLF10) contributes to distant migration of PDAC. The role of KLF10 in modulating tumorigenesis and stem cell phenotypes of PDAC is unclear. METHODS Additional depletion of KLF10 in KC (LSL: KrasG12D; Pdx1-Cre) mice, a spontaneous murine PDAC model, was established to evaluate tumorigenesis. Tumor specimens of PDAC patients were immune-stained of KLF10 to correlate with local recurrence after curative resection. Conditional overexpressing KLF10 in MiaPaCa and stably depleting KLF10 in Panc-1 (Panc-1-pLKO-shKLF10) cells were established for evaluating sphere formation, stem cell markers expression and tumor growth. The signal pathways modulated by KLF10 for PDAC stem cell phenotypes were disclosed by microarray analysis and validated by western blot, qRT-PCR, luciferase reporter assay. Candidate targets to reverse PDAC tumor growth were demonstrated in murine model. RESULTS KLF10, deficient in two-thirds of 105 patients with resected pancreatic PDAC, was associated with rapid local recurrence and large tumor size. Additional KLF10 depletion in KC mice accelerated progression from pancreatic intraepithelial neoplasia to PDAC. Increased sphere formation, expression of stem cell markers, and tumor growth were observed in Panc-1-pLKO-shKLF10 compared with vector control. Genetically or pharmacologically overexpression of KLF10 reversed the stem cell phenotypes induced by KLF10 depletion. Ingenuity pathway analysis and gene set enrichment analysis showed that Notch signaling molecules, including Notch receptors 3 and 4, were over-expressed in Panc-1-pLKO-shKLF10. KLF10 transcriptionally suppressed Notch-3 and -4 by competing with E74-like ETS transcription factor 3, a positive regulator, for promoter binding. Downregulation of Notch signaling, either genetically or pharmacologically, ameliorated the stem cell phenotypes of Panc-1-pLKO-shKLF10. The combination of metformin, which upregulated KLF10 expression via phosphorylating AMPK, and evodiamine, a non-toxic Notch-3 methylation stimulator, delayed tumor growth of PDAC with KLF10 deficiency in mice without prominent toxicity. CONCLUSIONS These results demonstrated a novel signaling pathway by which KLF10 modulates stem cell phenotypes in PDAC through transcriptionally regulating Notch signaling pathway. The elevation of KLF10 and suppression of Notch signaling may jointly reduce PDAC tumorigenesis and malignant progression.
Collapse
Affiliation(s)
- Yi-Chih Tsai
- National Institute of Cancer Research, National Health Research Institutes, R1-2034, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Kung Hung Cheng
- National Institute of Cancer Research, National Health Research Institutes, R1-2034, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Shih Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, R1-2034, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shun En Chuang
- National Institute of Cancer Research, National Health Research Institutes, R1-2034, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Su Liang Chen
- National Institute of Cancer Research, National Health Research Institutes, R1-2034, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Tze-Sing Huang
- National Institute of Cancer Research, National Health Research Institutes, R1-2034, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Hui-Ju Ch'ang
- National Institute of Cancer Research, National Health Research Institutes, R1-2034, 35 Keyan Road, Zhunan, Miaoli County, 35053, Taiwan.
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
- Department of Oncology, School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
4
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
5
|
Li D, Zhang J, Cheng C, Hou K, Wang X, Zhu L, Li B, Du Z, Wang J, Wang J. Effects of ecotoxicity of penoxsulam single and co-exposure with AgNPs on Eisenia fetida. CHEMOSPHERE 2022; 307:136134. [PMID: 36028129 DOI: 10.1016/j.chemosphere.2022.136134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
Penoxsulam (PNX) and silver nanoparticles (AgNPs) are likely to coexist in soils due to continuous use. However, the ecotoxicity of PNX in earthworms and the effect of AgNPs on PNX toxicity are unknown. Therefore, the toxicity of PNX (0.05, 0.5, and 2.5 mg/kg) single and co-exposure with AgNPs (10 mg/kg) after 28 and 56 days on Eisenia fetida (E. fetida) was investigated from biochemical, genetic, histopathological, and transcriptomic aspects. The results showed that the low concentration of PNX (0.05PNX) had almost no effect on the biochemical level of E. fetida. However, the addition of AgNPs resulted in 0.05PNX causing E. fetida to produce excessive reactive oxygen species, and the activity of antioxidant and detoxification enzymes were interfered, resulting in lipid peroxidation and DNA damage. From the genetic level, even the lowest concentration of PNX can significantly interfere with the expression of functional genes, thus inducing oxidative stress and apoptosis and inhibited reproductive behavior in E. fetida. The integrated biomarker response results at the biochemical and genetic levels showed that the comprehensive toxicity of PNX and PNX + AgNPs on E. fetida was PNX dose-dependent. And the toxicity of all co-exposure groups was greater than that of the PNX only exposure groups. Furthermore, the addition of AgNPs significantly increased the damage of PNX on E. fetida intestinal tissue. Meanwhile, transcriptomic analysis showed that PNX + AgNPs had a greater effect on E. fetida than PNX single, and multiple pathways related to oxidative stress, inflammation, and cellular process regulation were disturbed. These results provide a basis for comprehensive evaluation of the ecotoxicity of PNX and confirm that the AgNPs does increase the ecotoxicity of PNX in soil environment.
Collapse
Affiliation(s)
- Dengtan Li
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Jingwen Zhang
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Chao Cheng
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Kaixuan Hou
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Xiaole Wang
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Lusheng Zhu
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Bing Li
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Zhongkun Du
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Jinhua Wang
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| | - Jun Wang
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, China.
| |
Collapse
|
6
|
Urbański A, Johnston P, Bittermann E, Keshavarz M, Paris V, Walkowiak-Nowicka K, Konopińska N, Marciniak P, Rolff J. Tachykinin-related peptides modulate immune-gene expression in the mealworm beetle Tenebrio molitor L. Sci Rep 2022; 12:17277. [PMID: 36241888 PMCID: PMC9568666 DOI: 10.1038/s41598-022-21605-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 09/29/2022] [Indexed: 01/06/2023] Open
Abstract
Tachykinins (TKs) are a group of conserved neuropeptides. In insects, tachykinin-related peptides (TRPs) are important modulators of several functions such as nociception and lipid metabolism. Recently, it has become clear that TRPs also play a role in regulating the insect immune system. Here, we report a transcriptomic analysis of changes in the expression levels of immune-related genes in the storage pest Tenebrio molitor after treatment with Tenmo-TRP-7. We tested two concentrations (10-8 and 10-6 M) at two time points, 6 and 24 h post-injection. We found significant changes in the transcript levels of a wide spectrum of immune-related genes. Some changes were observed 6 h after the injection of Tenmo-TRP-7, especially in relation to its putative anti-apoptotic action. Interestingly, 24 h after the injection of 10-8 M Tenmo-TRP-7, most changes were related to the regulation of the cellular response. Applying 10-6 M Tenmo-TRP-7 resulted in the downregulation of genes associated with humoral responses. Injecting Tenmo-TRP-7 did not affect beetle survival but led to a reduction in haemolymph lysozyme-like antibacterial activity, consistent with the transcriptomic data. The results confirmed the immunomodulatory role of TRP and shed new light on the functional homology between TRPs and TKs.
Collapse
Affiliation(s)
- Arkadiusz Urbański
- grid.5633.30000 0001 2097 3545Department of Animal Physiology and Developmental Biology, Adam Mickiewicz University, Poznań, Poland ,grid.14095.390000 0000 9116 4836Evolutionary Biology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Paul Johnston
- Berlin Centre for Genomics in Biodiversity Research, Berlin, Germany ,grid.419247.d0000 0001 2108 8097Leibniz-Institute of Freshwater Ecology and Inland Fisheries (IGB), Berlin, Germany
| | - Elisa Bittermann
- grid.14095.390000 0000 9116 4836Evolutionary Biology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Maryam Keshavarz
- grid.14095.390000 0000 9116 4836Evolutionary Biology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Véronique Paris
- grid.14095.390000 0000 9116 4836Evolutionary Biology, Institute for Biology, Freie Universität Berlin, Berlin, Germany ,grid.1008.90000 0001 2179 088XBio 21 Institute, University of Melbourne, Parkville, VIC 3052 Australia
| | - Karolina Walkowiak-Nowicka
- grid.5633.30000 0001 2097 3545Department of Animal Physiology and Developmental Biology, Adam Mickiewicz University, Poznań, Poland
| | - Natalia Konopińska
- grid.5633.30000 0001 2097 3545Department of Animal Physiology and Developmental Biology, Adam Mickiewicz University, Poznań, Poland
| | - Paweł Marciniak
- grid.5633.30000 0001 2097 3545Department of Animal Physiology and Developmental Biology, Adam Mickiewicz University, Poznań, Poland
| | - Jens Rolff
- grid.14095.390000 0000 9116 4836Evolutionary Biology, Institute for Biology, Freie Universität Berlin, Berlin, Germany ,grid.452299.1Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB), Berlin, Germany
| |
Collapse
|
7
|
Rahman MM, Sarker MT, Alam Tumpa MA, Yamin M, Islam T, Park MN, Islam MR, Rauf A, Sharma R, Cavalu S, Kim B. Exploring the recent trends in perturbing the cellular signaling pathways in cancer by natural products. Front Pharmacol 2022; 13:950109. [PMID: 36160435 PMCID: PMC9498834 DOI: 10.3389/fphar.2022.950109] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is commonly thought to be the product of irregular cell division. According to the World Health Organization (WHO), cancer is the major cause of death globally. Nature offers an abundant supply of bioactive compounds with high therapeutic efficacy. Anticancer effects have been studied in a variety of phytochemicals found in nature. When Food and Drug Administration (FDA)-approved anticancer drugs are combined with natural compounds, the effectiveness improves. Several agents have already progressed to clinical trials based on these promising results of natural compounds against various cancer forms. Natural compounds prevent cancer cell proliferation, development, and metastasis by inducing cell cycle arrest, activating intrinsic and extrinsic apoptosis pathways, generating reactive oxygen species (ROS), and down-regulating activated signaling pathways. These natural chemicals are known to affect numerous important cellular signaling pathways, such as NF-B, MAPK, Wnt, Notch, Akt, p53, AR, ER, and many others, to cause cell death signals and induce apoptosis in pre-cancerous or cancer cells without harming normal cells. As a result, non-toxic “natural drugs” taken from nature’s bounty could be effective for the prevention of tumor progression and/or therapy of human malignancies, either alone or in combination with conventional treatments. Natural compounds have also been shown in preclinical studies to improve the sensitivity of resistant cancers to currently available chemotherapy agents. To summarize, preclinical and clinical findings against cancer indicate that natural-sourced compounds have promising anticancer efficacy. The vital purpose of these studies is to target cellular signaling pathways in cancer by natural compounds.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Taslim Sarker
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mst. Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Yamin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Tamanna Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Pakistan
- *Correspondence: Abdur Rauf, ; Bonglee Kim,
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Abdur Rauf, ; Bonglee Kim,
| |
Collapse
|
8
|
Sharma D, Bisen S, Kaur G, Van Buren EC, Rao GN, Singh NK. IL-33 enhances Jagged1 mediated NOTCH1 intracellular domain (NICD) deubiquitination and pathological angiogenesis in proliferative retinopathy. Commun Biol 2022; 5:479. [PMID: 35589941 PMCID: PMC9120174 DOI: 10.1038/s42003-022-03432-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/29/2022] [Indexed: 01/10/2023] Open
Abstract
Pathological retinal neovascularization (NV) is a clinical manifestation of various proliferative retinopathies, and treatment of NV using anti-VEGF therapies is not selective, as it also impairs normal retinal vascular growth and function. Here, we show that genetic deletion or siRNA-mediated downregulation of IL-33 reduces pathological NV in a murine model of oxygen-induced retinopathy (OIR) with no effect on the normal retinal repair. Furthermore, our fluorescent activated cell sorting (FACS) data reveals that the increase in IL-33 expression is in endothelial cells (ECs) of the hypoxic retina and conditional genetic deletion of IL-33 in retinal ECs reduces pathological NV. In vitro studies using human retinal microvascular endothelial cells (HRMVECs) show that IL-33 induces sprouting angiogenesis and requires NFkappaB-mediated Jagged1 expression and Notch1 activation. Our data also suggest that IL-33 enhances de-ubiquitination and stabilization of Notch1 intracellular domain via its interaction with BRCA1-associated protein 1 (BAP1) and Numb in HRMVECs and a murine model of OIR.
Collapse
Affiliation(s)
- Deepti Sharma
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Shivantika Bisen
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Geetika Kaur
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Eric C Van Buren
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nikhlesh K Singh
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA.
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48202, USA.
| |
Collapse
|
9
|
Zhu I, Liu R, Garcia JM, Hyrenius-Wittsten A, Piraner DI, Alavi J, Israni DV, Liu B, Khalil AS, Roybal KT. Modular design of synthetic receptors for programmed gene regulation in cell therapies. Cell 2022; 185:1431-1443.e16. [PMID: 35427499 PMCID: PMC9108009 DOI: 10.1016/j.cell.2022.03.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 01/20/2022] [Accepted: 03/15/2022] [Indexed: 01/01/2023]
Abstract
Synthetic biology has established powerful tools to precisely control cell function. Engineering these systems to meet clinical requirements has enormous medical implications. Here, we adopted a clinically driven design process to build receptors for the autonomous control of therapeutic cells. We examined the function of key domains involved in regulated intramembrane proteolysis and showed that systematic modular engineering can generate a class of receptors that we call synthetic intramembrane proteolysis receptors (SNIPRs) that have tunable sensing and transcriptional response abilities. We demonstrate the therapeutic potential of the receptor platform by engineering human primary T cells for multi-antigen recognition and production of dosed, bioactive payloads relevant to the treatment of disease. Our design framework enables the development of fully humanized and customizable transcriptional receptors for the programming of therapeutic cells suitable for clinical translation.
Collapse
Affiliation(s)
- Iowis Zhu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
| | - Raymond Liu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
| | - Julie M Garcia
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
| | - Axel Hyrenius-Wittsten
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA; Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund 221 84, Sweden
| | - Dan I Piraner
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
| | - Josef Alavi
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
| | - Divya V Israni
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Bin Liu
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA; Department of Anesthesia, University of California, San Francisco, San Francisco, CA 94110, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ahmad S Khalil
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA 02215, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Kole T Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Gladstone UCSF Institute for Genetic Immunology, San Francisco, CA 94107, USA; UCSF Cell Design Institute, San Francisco, CA 94158, USA.
| |
Collapse
|
10
|
Schnute B, Shimizu H, Lyga M, Baron M, Klein T. Ubiquitylation is required for the incorporation of the Notch receptor into intraluminal vesicles to prevent prolonged and ligand-independent activation of the pathway. BMC Biol 2022; 20:65. [PMID: 35264151 PMCID: PMC8908686 DOI: 10.1186/s12915-022-01245-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ubiquitylation of the ligands and the receptor plays an important part in the regulation of the activity of the evolutionary conserved Notch signalling pathway. However, its function for activation of Notch is not completely understood, despite the identification of several E3 ligases devoted to the receptor. RESULTS Here we analysed a variant of the Notch receptor where all lysines in its intracellular domain are replaced by arginines. Our analysis of this variant revealed that ubiquitylation of Notch is not essential for its endocytosis. We identified two functions for ubiquitylation of lysines in the Notch receptor. First, it is required for the degradation of free Notch intracellular domain (NICD) in the nucleus, which prevents a prolonged activation of the pathway. More importantly, it is also required for the incorporation of Notch into intraluminal vesicles of maturing endosomes to prevent ligand-independent activation of the pathway from late endosomal compartments. CONCLUSIONS The findings clarify the role of lysine-dependent ubiquitylation of the Notch receptor and indicate that Notch is endocytosed by several independent operating mechanisms.
Collapse
Affiliation(s)
- Björn Schnute
- Institute of Genetics, Heinrich-Heine-University Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Hideyuki Shimizu
- School of Biological Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Marvin Lyga
- Institute of Genetics, Heinrich-Heine-University Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Martin Baron
- School of Biological Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-University Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany.
| |
Collapse
|
11
|
Tang X, Xiao X, Sun H, Zheng S, Xiao X, Guo Z, Liu X, Sun W. 96DRA-urine: A high throughput sample preparation method for urinary proteome analysis. J Proteomics 2022; 257:104529. [PMID: 35181559 DOI: 10.1016/j.jprot.2022.104529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 01/25/2022] [Accepted: 02/13/2022] [Indexed: 11/26/2022]
Abstract
Mass spectrometry (MS)-based urinary proteomics is increasingly used for clinical research. A critical step in urinary proteomic analysis comprises the implementation of a reliable sample preparation method with high yields of peptides and proteins. In this study, we developed a urinary sample preparation method, DRA-Urine (Direct reduction/alkylation in urine), which urinary proteins were directly reduced/alkylated in urine, and then precipitated by acetone, washed and digestion on an ultrafilter unit. The qualitative and quantitative comparison of different urinary sample preparation methods (in-solution methods and ultrafilter-assisted methods) showed that DRA-Urine could achieve better results. Adapting DRA-Urine protocol to a 96-well format, namely 96DRA-Urine, shortened the time for buffer change and improved sample preparation throughput. The results showed that 96DRA-Urine displayed similar proteomic performance to DRA-Urine. Finally, the 96DRA-Urine method was used in a label-free, small pilot biomarker discovery analysis for differential urinary proteome analysis of bladder cancer urine. The results showed that urinary proteins could differentiate bladder cancer (BCa) patients from healthy controls and distinguish high-grade BCa from low-grade BCa with area under the curve (AUC) values of 0.972 and 0.847, respectively. Consequently, the 96DRA-Urine method might be a high-throughput method for preparing body fluid samples used in clinical research but needs to be further verified.
Collapse
Affiliation(s)
- Xiaoyue Tang
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China; Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaoping Xiao
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China; Cytology Lab, Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Haidan Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China
| | - Shuxin Zheng
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China
| | - Xiaolian Xiao
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China
| | - Zhengguang Guo
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China
| | - Xiaoyan Liu
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China
| | - Wei Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, China.
| |
Collapse
|
12
|
Zada S, Hwang JS, Lai TH, Pham TM, Ahmed M, Elashkar O, Kim W, Kim DR. Autophagy-mediated degradation of NOTCH1 intracellular domain controls the epithelial to mesenchymal transition and cancer metastasis. Cell Biosci 2022; 12:17. [PMID: 35164848 PMCID: PMC8842742 DOI: 10.1186/s13578-022-00752-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
Backgound Autophagy controls levels of cellular components during normal and stress conditions; thus, it is a pivotal process for the maintenance of cell homeostasis. In cancer, autophagy protects cells from cancerous transformations that can result from genomic instability induced by reactive oxygen species or other damaged components, but it can also promote cancer survival by providing essential nutrients during the metabolic stress condition of cancer progression. However, the molecular mechanism underlying autophagy-dependent regulation of the epithelial to mesenchymal transition (EMT) and metastasis is still elusive. Methods The intracellular level of NOTCH1 intracellular domain (NICD) in several cancer cells was studied under starvation, treatment with chloroquine or ATG7-knockdown. The autophagy activity in these cells was assessed by immunocytochemistry and molecular analyses. Cancer cell migration and invasion under modulation of autophagy were determined by in vitro scratch and Matrigel assays. Results In the study, autophagy activation stimulated degradation of NICD, a key transcriptional regulator of the EMT and cancer metastasis. We also found that NICD binds directly to LC3 and that the NICD/LC3 complex associates with SNAI1 and sequestosome 1 (SQSTM1)/p62 proteins. Furthermore, the ATG7 knockdown significantly inhibited degradation of NICD under starvation independent of SQSTM1-associated proteasomal degradation. In addition, NICD degradation by autophagy associated with the cellular level of SNAI1. Indeed, autophagy inhibited nuclear translocation of NICD protein and consequently decreased the transcriptional activity of its target genes. Autophagy activation substantially suppressed in vitro cancer cell migration and invasion. We also observed that NICD and SNAI1 levels in tissues from human cervical and lung cancer patients correlated inversely with expression of autophagy-related proteins. Conclusions These findings suggest that the cellular level of NICD is regulated by autophagy during cancer progression and that targeting autophagy-dependent NICD/SNAI1 degradation could be a strategy for the development of cancer therapeutics. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00752-3.
Collapse
Affiliation(s)
- Sahib Zada
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea.,Cancer Biology and Immunology Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Trang Minh Pham
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Omar Elashkar
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Wanil Kim
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea.
| |
Collapse
|
13
|
Ge Y, Wang J, Zhang H, Li J, Ye M, Jin X. Fate of hematopoietic stem cells determined by Notch1 signaling (Review). Exp Ther Med 2022; 23:170. [PMID: 35069851 PMCID: PMC8764575 DOI: 10.3892/etm.2021.11093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/17/2021] [Indexed: 11/05/2022] Open
Abstract
Regulation of the fate of hematopoietic stem cells (HSCs), including silencing, self-renewal or differentiation into blood line cells, is crucial to maintain the homeostasis of the human blood system and prevent leukemia. Notch1, a key receptor in the Notch signaling pathway, plays an important regulatory role in these properties of HSCs, particularly in the maintenance of the stemness of HSCs. In recent decades, the ubiquitination modification of Notch1 has been gradually revealed, and also demonstrated to affect the proliferation and differentiation of HSCs. Therefore, a detailed elucidation of Notch1 and its ubiquitination modification may help to improve understanding of the maintenance of HSC properties and the pathogenesis of leukemia. In addition, it may aid in identifying potential therapeutic targets for specific leukemias and provide potential prognostic indicators for HSC transplantation (HSCT). In the present review, the association between Notch1 and HSCs and the link between the ubiquitination modification of Notch1 and HSCs were described. In addition, the association between abnormal HSCs mediated by Notch1 or ubiquitinated Notch1and T-cell acute lymphoblastic leukemia (T-ALL) was also examined, which provides a promising direction for clinical application.
Collapse
Affiliation(s)
- Yidong Ge
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jie Wang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Hui Zhang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jinyun Li
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Meng Ye
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaofeng Jin
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
14
|
Zhang F, Chen Y, Shen J, Zhang J. The Ubiquitin Conjugating Enzyme UbcD1 is Required for Notch Signaling Activation During Drosophila Wing Development. Front Genet 2021; 12:770853. [PMID: 34712275 PMCID: PMC8546230 DOI: 10.3389/fgene.2021.770853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Notch signaling pathway plays crucial roles in animal development. Protein ubiquitination contributes to Notch signaling regulation by governing the stability and activity of major signaling components. Studies in Drosophila have identified multiple ubiquitin ligases and deubiquitinating enzymes that modify Notch ligand and receptor proteins. The fate of ubiquitinated substrates depend on topologies of the attached ubiquitin chains, which are determined by the ubiquitin conjugating enzymes (E2 enzymes). However, which E2 enzymes participate in Notch signal transduction remain elusive. Here, we report that the E2 enzyme UbcD1 is required for Notch signaling activation during Drosophila wing development. Mutations of UbcD1 lead to marginal nicks in the adult wing and reduction of Notch signaling targets expression in the wing imaginal disc. Genetic analysis reveal that UbcD1 functions in the signaling receiving cells prior to cleavage of the Notch protein. We provide further evidence suggesting that UbcD1 is likely involved in endocytic trafficking of Notch protein. Our results demonstrate that UbcD1 positively regulates Notch signaling and thus reveal a novel role of UbcD1 in development.
Collapse
Affiliation(s)
- Fengchao Zhang
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yao Chen
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Shen
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Junzheng Zhang
- MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Martinez Lyons A, Boulter L. The developmental origins of Notch-driven intrahepatic bile duct disorders. Dis Model Mech 2021; 14:dmm048413. [PMID: 34549776 PMCID: PMC8480193 DOI: 10.1242/dmm.048413] [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] [Indexed: 11/30/2022] Open
Abstract
The Notch signaling pathway is an evolutionarily conserved mechanism of cell-cell communication that mediates cellular proliferation, cell fate specification, and maintenance of stem and progenitor cell populations. In the vertebrate liver, an absence of Notch signaling results in failure to form bile ducts, a complex tubular network that radiates throughout the liver, which, in healthy individuals, transports bile from the liver into the bowel. Loss of a functional biliary network through congenital malformations during development results in cholestasis and necessitates liver transplantation. Here, we examine to what extent Notch signaling is necessary throughout embryonic life to initiate the proliferation and specification of biliary cells and concentrate on the animal and human models that have been used to define how perturbations in this signaling pathway result in developmental liver disorders.
Collapse
Affiliation(s)
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
| |
Collapse
|
16
|
Ubiquitination and Deubiquitination in Oral Disease. Int J Mol Sci 2021; 22:ijms22115488. [PMID: 34070986 PMCID: PMC8197098 DOI: 10.3390/ijms22115488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 01/07/2023] Open
Abstract
Oral health is an integral part of the general health and well-being of individuals. The presence of oral disease is potentially indicative of a number of systemic diseases and may contribute to their early diagnosis and treatment. The ubiquitin (Ub) system has been shown to play a role in cellular immune response, cellular development, and programmed cell death. Ubiquitination is a post-translational modification that occurs in eukaryotes. Its mechanism involves a number of factors, including Ub-activating enzymes, Ub-conjugating enzymes, and Ub protein ligases. Deubiquitinating enzymes, which are proteases that reversely modify proteins by removing Ub or Ub-like molecules or remodeling Ub chains on target proteins, have recently been regarded as crucial regulators of ubiquitination-mediated degradation and are known to significantly affect cellular pathways, a number of biological processes, DNA damage response, and DNA repair pathways. Research has increasingly shown evidence of the relationship between ubiquitination, deubiquitination, and oral disease. This review investigates recent progress in discoveries in diseased oral sites and discusses the roles of ubiquitination and deubiquitination in oral disease.
Collapse
|
17
|
Taira Y, Wada H, Hayashi S, Kageyama Y. polished rice mediates ecdysone-dependent control of Drosophila embryonic organogenesis. Genes Cells 2021; 26:269-281. [PMID: 33621395 DOI: 10.1111/gtc.12841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 01/08/2023]
Abstract
In many animals, progression of developmental stages is temporally controlled by steroid hormones. In Drosophila, the level of ecdysone titer oscillates and developmental stage transitions, such as larval molting and metamorphosis, are induced at each of ecdysone peaks. Ecdysone titer also peaks at the stage of mid-embryogenesis and the embryonic ecdysone is necessary for morphogenesis of several organs, although the regulatory mechanisms of embryonic organogenesis dependent on ecdysone signaling are still open questions. In this study, we find that absence or interruption of embryonic ecdysone signaling caused multiple defects in the tracheal system, including decrease in luminal protein deposition, uneven dilation of the dorsal trunk and loss of terminal branches. We also reveal that an ecdysone-inducible gene polished rice (pri) is essential for tip cell fate decision in dorsal branches. As over-expression of pri can restore the defects caused by disturbance of ecdysone biosynthesis, pri functions as one of the major mediators of embryonic ecdysone signal in tracheogenesis. These results demonstrate that ecdysone and its downstream target pri play essential roles in tracheal development by modulating cell fate decision.
Collapse
Affiliation(s)
- Yuki Taira
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan
| | - Housei Wada
- Laboratory for Morphogenetic Signaling, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Shigeo Hayashi
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan.,Laboratory for Morphogenetic Signaling, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yuji Kageyama
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan.,Biosignal Research Center, Kobe University, Kobe, Japan
| |
Collapse
|
18
|
Selenite Inhibits Notch Signaling in Cells and Mice. Int J Mol Sci 2021; 22:ijms22052518. [PMID: 33802299 PMCID: PMC7959125 DOI: 10.3390/ijms22052518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 02/05/2023] Open
Abstract
Selenium is an essential micronutrient with a wide range of biological effects in mammals. The inorganic form of selenium, selenite, is supplemented to relieve individuals with selenium deficiency and to alleviate associated symptoms. Additionally, physiological and supranutritional selenite have shown selectively higher affinity and toxicity towards cancer cells, highlighting their potential to serve as chemotherapeutic agents or adjuvants. At varying doses, selenite extensively regulates cellular signaling and modulates many cellular processes. In this study, we report the identification of Delta–Notch signaling as a previously uncharacterized selenite inhibited target. Our transcriptomic results in selenite treated primary mouse hepatocytes revealed that the transcription of Notch1, Notch2, Hes1, Maml1, Furin and c-Myc were all decreased following selenite treatment. We further showed that selenite can inhibit Notch1 expression in cultured MCF7 breast adenocarcinoma cells and HEPG2 liver carcinoma cells. In mice acutely treated with 2.5 mg/kg selenite via intraperitoneal injection, we found that Notch1 expression was drastically lowered in liver and kidney tissues by 90% and 70%, respectively. Combined, these results support selenite as a novel inhibitor of Notch signaling, and a plausible mechanism of inhibition has been proposed. This discovery highlights the potential value of selenite applied in a pathological context where Notch is a key drug target in diseases such as cancer, fibrosis, and neurodegenerative disorders.
Collapse
|
19
|
Dutta D, Sharma V, Mutsuddi M, Mukherjee A. Regulation of Notch signaling by E3 ubiquitin ligases. FEBS J 2021; 289:937-954. [PMID: 33644958 DOI: 10.1111/febs.15792] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/07/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022]
Abstract
Notch signaling is an evolutionarily conserved pathway that is widely used for multiple cellular events during development. Activation of the Notch pathway occurs when the ligand from a neighboring cell binds to the Notch receptor and induces cleavage of the intracellular domain of Notch, which further translocates into the nucleus to activate its downstream genes. The involvement of the Notch pathway in diverse biological events is possible due to the complexity in its regulation. In order to maintain tight spatiotemporal regulation, the Notch receptor, as well as its ligand, undergoes a series of physical and biochemical modifications that, in turn, helps in proper maintenance and fine-tuning of the signaling outcome. Ubiquitination is the post-translational addition of a ubiquitin molecule to a substrate protein, and the process is regulated by E3 ubiquitin ligases. The present review describes the involvement of different E3 ubiquitin ligases that play an important role in the regulation and maintenance of proper Notch signaling and how perturbation in ubiquitination results in abnormal Notch signaling leading to a number of human diseases.
Collapse
Affiliation(s)
- Debdeep Dutta
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
20
|
Gratton R, Tricarico PM, d’Adamo AP, Bianco AM, Moura R, Agrelli A, Brandão L, Zupin L, Crovella S. Notch Signaling Regulation in Autoinflammatory Diseases. Int J Mol Sci 2020; 21:E8847. [PMID: 33238371 PMCID: PMC7700323 DOI: 10.3390/ijms21228847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/13/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022] Open
Abstract
Notch pathway is a highly conserved intracellular signaling route that modulates a vast variety of cellular processes including proliferation, differentiation, migration, cell fate and death. Recently, the presence of a strict crosstalk between Notch signaling and inflammation has been described, although the precise molecular mechanisms underlying this interplay have not yet been fully unravelled. Disruptions in Notch cascade, due both to direct mutations and/or to an altered regulation in the core components of Notch signaling, might lead to hypo- or hyperactivation of Notch target genes and signaling molecules, ultimately contributing to the onset of autoinflammatory diseases. To date, alterations in Notch signaling have been reported as associated with three autoinflammatory disorders, therefore, suggesting a possible role of Notch in the pathogenesis of the following diseases: hidradenitis suppurativa (HS), Behçet disease (BD), and giant cell arteritis (GCA). In this review, we aim at better characterizing the interplay between Notch and autoinflammatory diseases, trying to identify the role of this signaling route in the context of these disorders.
Collapse
Affiliation(s)
- Rossella Gratton
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Paola Maura Tricarico
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Adamo Pio d’Adamo
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Anna Monica Bianco
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Ronald Moura
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Almerinda Agrelli
- Department of Pathology, Federal University of Pernambuco, Recife 50670-901, Brazil;
| | - Lucas Brandão
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Luisa Zupin
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.G.); (A.P.d.); (A.M.B.); (R.M.); (L.B.); (L.Z.)
| | - Sergio Crovella
- Department of Biological and Environmental Sciences, College of Arts and Sciences, University of Qatar, Doha 2713, Qatar;
| |
Collapse
|
21
|
Yoo SS, Lee JH, Hong MJ, Choi JE, Kang HG, Do SK, Kim JH, Baek SA, Choi SH, Lee WK, Lee YH, Seo H, Lee J, Lee SY, Cha SI, Kim CH, Park JY. Effect of genetic variation in Notch regulator DTX1 on SCLC prognosis compared with the effect on NSCLC prongosis. Thorac Cancer 2020; 11:2698-2703. [PMID: 32700476 PMCID: PMC7471053 DOI: 10.1111/1759-7714.13566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/30/2022] Open
Abstract
Deltex-1 (DTX1) is a negative regulator of the Notch signaling pathway. Here, we investigated the clinical effect of DTX1 rs1732786A > G, which is associated with better prognosis in patients with early-stage non-small cell lung cancer (NSCLC), in 261 patients with small cell lung cancer (SCLC). DTX1 rs1732786A > G was associated with a significantly worse chemotherapy response and lower overall survival in the codominant model (odds ratio = 0.42, 95% confidence interval [CI]: 0.26-0.66, P = 2 × 10-4 ; hazard ratio = 1.47, 95% CI: 1.17-1.84, P = 0.001, respectively). An in vitro luciferase assay was performed, and the 1732786G allele demonstrated significantly higher promoter activity than the 1732786A allele (P = 2 × 10-7 ). In summary, DTX1 rs1732786A > G was associated with poor prognosis in patients with SCLC as opposed to patients with NSCLC. KEY POINTS: SIGNIFICANT FINDINGS OF THE STUDY: DTX1 rs1732786A > G was associated with better prognosis in patients with early-stage non-small cell lung cancer (NSCLC) in our previous study. WHAT THIS STUDY ADDS: DTX1 rs1732786A > G was associated with a significantly worse chemotherapy response and lower overall survival in small cell lung cancer (SCLC).
Collapse
Affiliation(s)
- Seung Soo Yoo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jang Hyuck Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Mi Jeong Hong
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Eun Choi
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo-Gyoung Kang
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sook Kyung Do
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Tumor Heterogeneity and Network (THEN) Research Center, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Hyun Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sun Ah Baek
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sun Ha Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Won Kee Lee
- Biostatistics, Medical Research Collaboration Center, Kyungpook National University, Daegu, Republic of Korea
| | - Yong Hoon Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hyewon Seo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jaehee Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Seung Ick Cha
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Chang Ho Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jae Yong Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.,Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
22
|
Luo Z, Mu L, Zheng Y, Shen W, Li J, Xu L, Zhong B, Liu Y, Zhou Y. NUMB enhances Notch signaling by repressing ubiquitination of NOTCH1 intracellular domain. J Mol Cell Biol 2020; 12:345-358. [PMID: 31504682 PMCID: PMC7288735 DOI: 10.1093/jmcb/mjz088] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/05/2019] [Accepted: 07/31/2019] [Indexed: 01/02/2023] Open
Abstract
The release and nuclear translocation of the intracellular domain of Notch receptor (NICD) is the prerequisite for Notch signaling-mediated transcriptional activation. NICD is subjected to various posttranslational modifications including ubiquitination. Here, we surprisingly found that NUMB proteins stabilize the intracellular domain of NOTCH1 receptor (N1ICD) by regulating the ubiquitin-proteasome machinery, which is independent of NUMB's role in modulating endocytosis. BAP1, a deubiquitinating enzyme (DUB), was further identified as a positive N1ICD regulator, and NUMB facilitates the association between N1ICD and BAP1 to stabilize N1ICD. Intriguingly, BAP1 stabilizes N1ICD independent of its DUB activity but relying on the BRCA1-inhibiting function. BAP1 strengthens Notch signaling and maintains stem-like properties of cortical neural progenitor cells. Thus, NUMB enhances Notch signaling by regulating the ubiquitinating activity of the BAP1-BRCA1 complex.
Collapse
Affiliation(s)
- Zhiyuan Luo
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Lili Mu
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Yue Zheng
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Wenchen Shen
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Jiali Li
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Lichao Xu
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Bo Zhong
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Ying Liu
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| | - Yan Zhou
- College of Life Sciences, Renmin Hospital of Wuhan University, Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430072, China
| |
Collapse
|
23
|
Dayekh K, Mequanint K. Comparative Studies of Fibrin-Based Engineered Vascular Tissues and Notch Signaling from Progenitor Cells. ACS Biomater Sci Eng 2020; 6:2696-2706. [DOI: 10.1021/acsbiomaterials.0c00255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Khalil Dayekh
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B9, Canada
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B9, Canada
| |
Collapse
|
24
|
Regulation of Notch Signaling in Drosophila melanogaster: The Role of the Heterogeneous Nuclear Ribonucleoprotein Hrp48 and Deltex. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1227:95-105. [DOI: 10.1007/978-3-030-36422-9_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Hosokawa K, Ishimaru H, Watanabe T, Fujimuro M. The Lysosome Pathway Degrades CD81 on the Cell Surface by Poly-ubiquitination and Clathrin-Mediated Endocytosis. Biol Pharm Bull 2019; 43:540-545. [PMID: 31902824 DOI: 10.1248/bpb.b19-01097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CD81 is a highly conserved four-transmembrane protein in mammals and widely expressed on many tissues. It belongs to the tetraspanin family and forms complexes with various cell surface membrane proteins. It also functions in cell migration and B-cell activation, which is induced by CD81 complexing with CD19, CD21 and the B-cell receptor. Thus, CD81 is thought to play a key role in regulating cell function and fate. However, little is known about the degradation mechanism of CD81. Here we found that CD81 on the plasma membrane is degraded by the lysosome pathway via endocytosis. The expression levels of CD81 in HEK293T cells treated with a proteasome inhibitor (lactacystin) and lysosome inhibitors (chloroquine and bafilomycin A1) were analyzed by flow cytometry. The expression of CD81 on the cell surface was increased by the lysosome inhibitors, but not lactacystin. A pulldown assay revealed that CD81 was conjugated with a K63- and K29-linked poly-ubiquitin chain before its degradation, and the poly-ubiquitination site was Lys8 at the N-terminal intracellular domain of CD81. Furthermore, mutant CD81, in which Lys8 was substituted with alanine (Ala), extended the CD81 half-life compared with wildtype. CD81 was mainly localized on the plasma membrane in normal cells, but also co-localized with lysosomal LAMP1 and early endosomal EEA1 in chloroquine-treated cells. Furthermore, a clathrin-mediated endocytosis inhibitor, chlorpromazine, stabilized CD81 expression on the cell surface. Hence, we demonstrated that CD81 is internalized by clathrin-mediated endocytosis and subsequently degraded via a lysosome pathway requiring the K63- and K29-linked poly-ubiquitination of CD81.
Collapse
Affiliation(s)
- Kohei Hosokawa
- Department of Cell Biology, Kyoto Pharmaceutical University
| | | | | | | |
Collapse
|
26
|
Lee JH, Shin KM, Lee SY, Hong MJ, Choi JE, Kang HG, Do SK, Lee WK, Lee EB, Seok Y, Jeong JY, Yoo SS, Lee J, Cha SI, Kim CH, Cho S, Jheon S, Kim YC, Oh IJ, Na KJ, Kim MS, Lee JM, Yang HC, Jung CY, Park CK, Lee MK, Kim DK, Park JY. Genetic Variant of Notch Regulator DTX1 Predicts Survival After Lung Cancer Surgery. Ann Surg Oncol 2019; 26:3756-3764. [DOI: 10.1245/s10434-019-07614-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Indexed: 08/30/2023]
|
27
|
Shin S, Kim K, Kim HR, Ylaya K, Do SI, Hewitt SM, Park HS, Roe JS, Chung JY, Song J. Deubiquitylation and stabilization of Notch1 intracellular domain by ubiquitin-specific protease 8 enhance tumorigenesis in breast cancer. Cell Death Differ 2019; 27:1341-1354. [PMID: 31527799 DOI: 10.1038/s41418-019-0419-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/31/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022] Open
Abstract
Notch, an essential factor in tissue development and homoeostasis, has been reported to play an oncogenic function in a variety of cancers. Here, we report ubiquitin-specific protease 8 (USP8) as a novel deubiquitylase of Notch1 intracellular domain (NICD). USP8 specifically stabilizes and deubiquitylates NICD through a direct interaction. The inhibition of USP8 downregulated the Notch signalling pathway via NICD destabilization, resulting in the retardation of cellular growth, wound closure, and colony forming ability of breast cancer cell lines. These phenomena were restored by the reconstitution of NICD or USP8, supporting the direct interaction between these two proteins. The expression levels of NICD and USP8 proteins were positively correlated in patients with advanced breast cancer. Taken together, our results suggest that USP8 functions as a positive regulator of Notch signalling, offering a therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Soyeon Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kyungeun Kim
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kris Ylaya
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sung-Im Do
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Republic of Korea
| | - Stephen M Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hee-Sae Park
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Joon-Yong Chung
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
28
|
Antfolk D, Antila C, Kemppainen K, Landor SKJ, Sahlgren C. Decoding the PTM-switchboard of Notch. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118507. [PMID: 31301363 PMCID: PMC7116576 DOI: 10.1016/j.bbamcr.2019.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/03/2019] [Accepted: 07/06/2019] [Indexed: 01/08/2023]
Abstract
The developmentally indispensable Notch pathway exhibits a high grade of pleiotropism in its biological output. Emerging evidence supports the notion of post-translational modifications (PTMs) as a modus operandi controlling dynamic fine-tuning of Notch activity. Although, the intricacy of Notch post-translational regulation, as well as how these modifications lead to multiples of divergent Notch phenotypes is still largely unknown, numerous studies show a correlation between the site of modification and the output. These include glycosylation of the extracellular domain of Notch modulating ligand binding, and phosphorylation of the PEST domain controlling half-life of the intracellular domain of Notch. Furthermore, several reports show that multiple PTMs can act in concert, or compete for the same sites to drive opposite outputs. However, further investigation of the complex PTM crosstalk is required for a complete understanding of the PTM-mediated Notch switchboard. In this review, we aim to provide a consistent and up-to-date summary of the currently known PTMs acting on the Notch signaling pathway, their functions in different contexts, as well as explore their implications in physiology and disease. Furthermore, we give an overview of the present state of PTM research methodology, and allude to a future with PTM-targeted Notch therapeutics.
Collapse
Affiliation(s)
- Daniel Antfolk
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Christian Antila
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Kati Kemppainen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Sebastian K-J Landor
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland; Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
29
|
Carrieri FA, Murray PJ, Ditsova D, Ferris MA, Davies P, Dale JK. CDK1 and CDK2 regulate NICD1 turnover and the periodicity of the segmentation clock. EMBO Rep 2019; 20:e46436. [PMID: 31267714 PMCID: PMC6607002 DOI: 10.15252/embr.201846436] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 03/11/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022] Open
Abstract
All vertebrates share a segmented body axis. Segments form from the rostral end of the presomitic mesoderm (PSM) with a periodicity that is regulated by the segmentation clock. The segmentation clock is a molecular oscillator that exhibits dynamic clock gene expression across the PSM with a periodicity that matches somite formation. Notch signalling is crucial to this process. Altering Notch intracellular domain (NICD) stability affects both the clock period and somite size. However, the mechanism by which NICD stability is regulated in this context is unclear. We identified a highly conserved site crucial for NICD recognition by the SCF E3 ligase, which targets NICD for degradation. We demonstrate both CDK1 and CDK2 can phosphorylate NICD in the domain where this crucial residue lies and that NICD levels vary in a cell cycle-dependent manner. Inhibiting CDK1 or CDK2 activity increases NICD levels both in vitro and in vivo, leading to a delay of clock gene oscillations and an increase in somite size.
Collapse
Affiliation(s)
- Francesca Anna Carrieri
- Division of Cell and Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| | | | - Dimitrinka Ditsova
- Division of Cell and Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| | | | - Paul Davies
- Medical Research Council Protein Phosphorylation and Ubiquitylation UnitSchool of Life SciencesUniversity of DundeeDundeeUK
| | - Jacqueline Kim Dale
- Division of Cell and Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| |
Collapse
|
30
|
Therapeutic Targeting of Notch Signaling Pathway in Hematological Malignancies. Mediterr J Hematol Infect Dis 2019; 11:e2019037. [PMID: 31308913 PMCID: PMC6613627 DOI: 10.4084/mjhid.2019.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/18/2019] [Indexed: 12/16/2022] Open
Abstract
The Notch pathway plays a key role in several processes, including stem-cell self-renewal, proliferation, and cell differentiation. Several studies identified recurrent mutations in hematological malignancies making Notch one of the most desirable targets in leukemia and lymphoma. The Notch signaling mediates resistance to therapy and controls cancer stem cells supporting the development of on-target therapeutic strategies to improve patients’ outcome. In this brief review, we outline the therapeutic potential of targeting Notch pathway in T-cell acute jlymphoblastic leukemia, chronic lymphocytic leukemia, and mantle cell lymphoma.
Collapse
|
31
|
Baldea I, Giurgiu L, Teacoe ID, Olteanu DE, Olteanu FC, Clichici S, Filip GA. Photodynamic Therapy in Melanoma - Where do we Stand? Curr Med Chem 2019; 25:5540-5563. [PMID: 29278205 DOI: 10.2174/0929867325666171226115626] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Malignant melanoma is one of the most aggressive malignant tumors, with unpredictable evolution. Despite numerous therapeutic options, like chemotherapy, BRAF inhibitors and immunotherapy, advanced melanoma prognosis remains severe. Photodynamic therapy (PDT) has been successfully used as the first line or palliative therapy for the treatment of lung, esophageal, bladder, non melanoma skin and head and neck cancers. However, classical PDT has shown some drawbacks that limit its clinical application in melanoma. OBJECTIVE The most important challenge is to overcome melanoma resistance, due to melanosomal trapping, presence of melanin, enhanced oxidative stress defense, defects in the apoptotic pathways, immune evasion, neoangiogenesis stimulation. METHOD In this review we considered: (1) main signaling molecular pathways deregulated in melanoma as potential targets for personalized therapy, including PDT, (2) results of the clinical studies regarding PDT of melanoma, especially advanced metastatic stage, (3) progresses made in the design of anti-melanoma photosensitizers (4) inhibition of tumor neoangiogenesis, as well as (5) advantages of the derived therapies like photothermal therapy, sonodynamic therapy. RESULTS PDT represents a promising alternative palliative treatment for advanced melanoma patients, mainly due to its minimal invasive character and low side effects. Efficient melanoma PDT requires: (1) improved, tumor targeted, NIR absorbing photosensitizers, capable of inducing high amounts of different ROS inside tumor and vasculature cells, possibly allowing a theranostic approach; (2) an efficient adjuvant immune therapy. CONCLUSION Combination of PDT with immune stimulation might be the key to overcome the melanoma resistance and to obtain better, sustainable clinical results.
Collapse
Affiliation(s)
- Ioana Baldea
- Physiology Department, University of Medicine and Pharmacy, Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Lorin Giurgiu
- Physiology Department, University of Medicine and Pharmacy, Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Ioana Diana Teacoe
- Physiology Department, University of Medicine and Pharmacy, Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Diana Elena Olteanu
- Physiology Department, University of Medicine and Pharmacy, Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Florin Catalin Olteanu
- Industrial Engineering and Management Department, Transylvania University, Brasov, Romania
| | - Simona Clichici
- Physiology Department, University of Medicine and Pharmacy, Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Gabriela Adriana Filip
- Physiology Department, University of Medicine and Pharmacy, Iuliu Hatieganu, Cluj-Napoca, Romania
| |
Collapse
|
32
|
Sorrentino C, Hossain F, Rodriguez PC, Sierra RA, Pannuti A, Osborne BA, Minter LM, Miele L, Morello S. Adenosine A2A Receptor Stimulation Inhibits TCR-Induced Notch1 Activation in CD8+T-Cells. Front Immunol 2019; 10:162. [PMID: 30792717 PMCID: PMC6374329 DOI: 10.3389/fimmu.2019.00162] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/18/2019] [Indexed: 12/23/2022] Open
Abstract
Notch receptors signaling is required for optimal T-cell activation and function. T-cell receptor (TCR) engagement can activate Notch receptors in T-cells in a ligand-independent fashion. In this study, we examined the role of adenosine A2A receptor (A2AR) signaling pathway in regulating the activity of Notch1 induced by TCR stimulation in CD8+T-cells. A selective A2AR agonist decreased Notch1 protein expression and Notch1 cleavage, and reduced transcripts of Notch1-target genes Hes1 and Myc in activated CD8+T-cells. Inhibition of TCR-induced Notch1 expression by an A2AR agonist was accompanied by increased cAMP concentration and mimicked by forskolin. This effect was associated with reduced IFN-γ and granzyme B production. The effect of an A2AR agonist was abrogated by a selective A2AR antagonist and absent in CD8+T-cells harvested from A2AR-/- mice. Stimulation of A2AR reduced Notch1 receptor levels by inhibiting upstream TCR signals, including ZAP70 phosphorylation, in turn impairing the generation of the active Notch1 intracellular domain (N1ICD). Direct activation of PKC with PMA and ionomycin bypassed A2AR-induced Notch1 inhibition. Overexpression of N1ICD in CD8+T-cells prevented the suppressive effects of an A2AR agonist on proliferation and cytokine release during activation. Our results identify the A2AR signaling pathway as an important regulator of TCR-induced Notch1 receptor activation in CD8+T-cells, and Notch as an important target of the immune suppressive effects of A2AR. We propose a mechanism whereby A2AR impairs CD8 T-cells function through inhibition of Notch1 receptor activation.
Collapse
Affiliation(s)
| | - Fokhrul Hossain
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | | | - Rosa A Sierra
- H. L. Moffitt Comprehensive Cancer Center, Tampa, FL, United States
| | - Antonio Pannuti
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Barbara A Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Lisa M Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Lucio Miele
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| |
Collapse
|
33
|
Estrogen-dependent DLL1-mediated Notch signaling promotes luminal breast cancer. Oncogene 2018; 38:2092-2107. [PMID: 30442981 PMCID: PMC6756232 DOI: 10.1038/s41388-018-0562-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 09/23/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022]
Abstract
Aberrant Notch signaling is implicated in several cancers, including breast cancer. However, the mechanistic details of the specific receptors and function of ligand-mediated Notch signaling that promote breast cancer remains elusive. In our studies we show that DLL1, a Notch signaling ligand, is significantly overexpressed in ERα+ luminal breast cancer. Intriguingly, DLL1 overexpression correlates with poor prognosis in ERα+ luminal breast cancer, but not in other subtypes of breast cancer. In addition, this effect is specific to DLL1, as other Notch ligands (DLL3, JAGGED1, and JAGGED2) do not influence the clinical outcome of ERα+ patients. Genetic studies show that DLL1-mediated Notch signaling in breast cancer is important for tumor cell proliferation, angiogenesis, and cancer stem cell function. Consistent with prognostic clinical data, we found the tumor-promoting function of DLL1 is exclusive to ERα+ luminal breast cancer, as loss of DLL1 inhibits both tumor growth and lung metastasis of luminal breast cancer. Importantly, we find that estrogen signaling stabilizes DLL1 protein by preventing its proteasomal and lysososmal degradations. Moreover, estrogen inhibits ubiquitination of DLL1. Together, our results highlight an unexpected and novel subtype-specific function of DLL1 in promoting luminal breast cancer that is regulated by estrogen signaling. Our studies also emphasize the critical role of assessing subtype-specific mechanisms driving tumor growth and metastasis to generate effective subtype-specific therapeutics.
Collapse
|
34
|
Zhang T, Guo L, Wang Y, Yang Y. Macroautophagy Regulates Nuclear NOTCH1 Activity Through Multiple p62 Binding Sites. IUBMB Life 2018; 70:985-994. [PMID: 30207627 DOI: 10.1002/iub.1891] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/17/2018] [Accepted: 05/30/2018] [Indexed: 01/07/2023]
Abstract
NOTCH1 is the prototype of the NOTCH family of single-pass transmembrane receptors and regulates many basic processes during embryonic development and human pathogenesis. In core to NOTCH1 activation are proteolytic cleavages that release its intracellular domain (NICD1), which in turn translocates to the nucleus to regulate gene transcription. Macroautophagy (hereafter autophagy) has been shown to promote the degradation of NOTCH1, but the underlying mechanisms remain elusive. Here, we show that autophagy promotes the degradation of NOTCH1 by p62-dependent binding between NICD1 and LC3, a component of the autophagosomes that execute autophagy. Strikingly, deleting any of the structural NICD1 domains fails to block the degradation of NICD1 by autophagy, and p62 binds to almost all these domains independently, indicating that p62 binds to multiple sites on NICD1 to promote its degradation. Intriguingly, inhibition of autophagy induces the accumulation of NICD1 in not only the cytoplasm but also the nucleus and increases the transcriptional activity of NICD1, and such regulation of nuclear NICD1 by autophagy is unique to NICD1 and not observed for all other NICDs (NICD2-4). Collectively, our results suggest that autophagy tightly controls nuclear NOTCH1 activity through multiple p62 binding sites, and that modulating autophagy activity may be useful for treating NOTCH1 related human diseases. © 2018 IUBMB Life, 70(10):985-994, 2018.
Collapse
Affiliation(s)
- Ting Zhang
- Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, College of Medicine and Science, Mayo Clinic, MN, USA
| | - Lixia Guo
- Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, College of Medicine and Science, Mayo Clinic, MN, USA
| | - Yuanyuan Wang
- Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, College of Medicine and Science, Mayo Clinic, MN, USA.,Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanan Yang
- Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, College of Medicine and Science, Mayo Clinic, MN, USA.,Developmental Therapeutics and Cell Biology Programs, Mayo Clinic Cancer Center, Mayo Clinic, MN, USA.,Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, MN, USA
| |
Collapse
|
35
|
Crosstalk between Notch, HIF-1α and GPER in Breast Cancer EMT. Int J Mol Sci 2018; 19:ijms19072011. [PMID: 29996493 PMCID: PMC6073901 DOI: 10.3390/ijms19072011] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022] Open
Abstract
The Notch signaling pathway acts in both physiological and pathological conditions, including embryonic development and tumorigenesis. In cancer progression, diverse mechanisms are involved in Notch-mediated biological responses, including angiogenesis and epithelial-mesenchymal-transition (EMT). During EMT, the activation of cellular programs facilitated by transcriptional repressors results in epithelial cells losing their differentiated features, like cell–cell adhesion and apical–basal polarity, whereas they gain motility. As it concerns cancer epithelial cells, EMT may be consequent to the evolution of genetic/epigenetic instability, or triggered by factors that can act within the tumor microenvironment. Following a description of the Notch signaling pathway and its major regulatory nodes, we focus on studies that have given insights into the functional interaction between Notch signaling and either hypoxia or estrogen in breast cancer cells, with a particular focus on EMT. Furthermore, we describe the role of hypoxia signaling in breast cancer cells and discuss recent evidence regarding a functional interaction between HIF-1α and GPER in both breast cancer cells and cancer-associated fibroblasts (CAFs). On the basis of these studies, we propose that a functional network between HIF-1α, GPER and Notch may integrate tumor microenvironmental cues to induce robust EMT in cancer cells. Further investigations are required in order to better understand how hypoxia and estrogen signaling may converge on Notch-mediated EMT within the context of the stroma and tumor cells interaction. However, the data discussed here may anticipate the potential benefits of further pharmacological strategies targeting breast cancer progression.
Collapse
|
36
|
Endocytic Trafficking of the Notch Receptor. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:99-122. [DOI: 10.1007/978-3-319-89512-3_6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
37
|
Man J, Yu X, Huang H, Zhou W, Xiang C, Huang H, Miele L, Liu Z, Bebek G, Bao S, Yu JS. Hypoxic Induction of Vasorin Regulates Notch1 Turnover to Maintain Glioma Stem-like Cells. Cell Stem Cell 2017; 22:104-118.e6. [PMID: 29198941 DOI: 10.1016/j.stem.2017.10.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/11/2017] [Accepted: 10/13/2017] [Indexed: 12/16/2022]
Abstract
Tumor hypoxia is associated with poor patient survival and is a characteristic of glioblastoma. Notch signaling is implicated in maintaining glioma stem-like cells (GSCs) within the hypoxic niche, although the molecular mechanisms linking hypoxia to Notch activation have not been clearly delineated. Here we show that Vasorin is a critical link between hypoxia and Notch signaling in GSCs. Vasorin is preferentially induced in GSCs by a HIF1α/STAT3 co-activator complex and stabilizes Notch1 protein at the cell membrane. This interaction prevents Numb from binding Notch1, rescuing it from Numb-mediated lysosomal degradation. Thus, Vasorin acts as a switch to augment Notch signaling under hypoxic conditions. Vasorin promotes tumor growth and reduces survival in mouse models of glioblastoma, and its expression correlates with increased aggression of human gliomas. These findings provide mechanistic insights into how hypoxia promotes Notch signaling in glioma and identify Vasorin as a potential therapeutic target.
Collapse
Affiliation(s)
- Jianghong Man
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA; National Center of Biomedical Analysis, Beijing 100850, China
| | - Xingjiang Yu
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA
| | - Haidong Huang
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA
| | - Wenchao Zhou
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA
| | - Chaomei Xiang
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA
| | - Haohao Huang
- National Center of Biomedical Analysis, Beijing 100850, China
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, Clinical Sciences Research Building, Room 657, 533 Bolivar Street, New Orleans, LA 70112, USA
| | - Zhenggang Liu
- Cell and Cancer Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Gurkan Bebek
- Department of Nutrition, Center for Proteomics and Bioinformatics, Case Western Reserve University, 10900 Euclid Avenue, BRB 921, Cleveland, OH 44106, USA
| | - Shideng Bao
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA; Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Jennifer S Yu
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE30, Cleveland, OH 44195, USA; Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA; Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, CA50, Cleveland, OH 44195, USA.
| |
Collapse
|
38
|
Ajima R, Suzuki E, Saga Y. Pofut1 point-mutations that disrupt O-fucosyltransferase activity destabilize the protein and abolish Notch1 signaling during mouse somitogenesis. PLoS One 2017; 12:e0187248. [PMID: 29095923 PMCID: PMC5667770 DOI: 10.1371/journal.pone.0187248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 10/17/2017] [Indexed: 01/20/2023] Open
Abstract
The segmental pattern of the vertebrate body is established via the periodic formation of somites from the presomitic mesoderm (PSM). This periodical process is controlled by the cyclic and synchronized activation of Notch signaling in the PSM. Protein O-fucosyltransferase1 (Pofut1), which transfers O-fucose to the EGF domains of the Notch1 receptor, is indispensable for Notch signaling activation. The Drosophila homologue Ofut1 was reported to control Notch localization via two different mechanisms, working as a chaperone for Notch or as a regulator of Notch endocytosis. However, these were found to be independent of O-fucosyltransferase activity because the phenotypes were rescued by Ofut1 mutants lacking O-fucosyltransferase activity. Pofut1 may also be involved in the Notch receptor localization in mice. However, the contribution of enzymatic activity of Pofut1 to the Notch receptor dynamics remains to be elucidated. In order to clarify the importance of the O-fucosyltransferase activity of Pofut1 for Notch signaling activation and the protein localization in the PSM, we established mice carrying point mutations at the 245th a.a. or 370-372th a.a., highly conserved amino-acid sequences whose mutations disrupt the O-fucosyltransferase activity of both Drosophila Ofut1 and mammalian Pofut1, with the CRISPR/Cas9 mediated genome-engineering technique. Both mutants displayed the same severely perturbed somite formation and Notch1 subcellular localization defects as the Pofut1 null mutants. In the mutants, Pofut1 protein, but not RNA, became undetectable by E9.5. Furthermore, both wild-type and mutant Pofut1 proteins were degraded through lysosome dependent machinery. Pofut1 protein loss in the point mutant embryos caused the same phenotypes as those observed in Pofut1 null embryos.
Collapse
Affiliation(s)
- Rieko Ajima
- Mammalian Development Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima, Shizuoka, Japan
- Mouse Research Supporting Unit, National Institute of Genetics, Mishima, Shizuoka, Japan
- Department of Genetics, SOKENDAI, Mishima, Shizuoka, Japan
- * E-mail: (RA); (YS)
| | - Emiko Suzuki
- Department of Genetics, SOKENDAI, Mishima, Shizuoka, Japan
- Gene Network Laboratory, Structural Biology Center, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Yumiko Saga
- Mammalian Development Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima, Shizuoka, Japan
- Mouse Research Supporting Unit, National Institute of Genetics, Mishima, Shizuoka, Japan
- Department of Genetics, SOKENDAI, Mishima, Shizuoka, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail: (RA); (YS)
| |
Collapse
|
39
|
Sapir T, Levy T, Kozer N, Shin I, Zamor V, Haffner-Krausz R, McGlade JC, Reiner O. Notch Activation by Shootin1 Opposing Activities on 2 Ubiquitin Ligases. Cereb Cortex 2017; 28:3115-3128. [DOI: 10.1093/cercor/bhx180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/23/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot, Israel
| | - Talia Levy
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot, Israel
| | - Noga Kozer
- Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 234 Herzl St., Rehovot, Israel
| | - Irina Shin
- Biological Services Unit, Weizmann Institute of Science, 234 Herzl St., Rehovot, Israel
| | - Vanessa Zamor
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot, Israel
| | - Rebecca Haffner-Krausz
- Department of Veterinary Resources, Weizmann Institute of Science, 234 Herzl St., Rehovot, Israel
| | - Jane C McGlade
- The Arthur and Sonia Labatt Brain Tumour Research Centre and Program in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON,Canada
- Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, ON, Canada
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, 234 Herzl St., Rehovot, Israel
| |
Collapse
|
40
|
|
41
|
Abstract
Newly synthesized transmembrane proteins undergo a series of steps to ensure that only the required amount of correctly folded protein is localized to the membrane. The regulation of protein quality and its abundance at the membrane are often controlled by ubiquitination, a multistep enzymatic process that results in the attachment of ubiquitin, or chains of ubiquitin to the target protein. Protein ubiquitination acts as a signal for sorting, trafficking, and the removal of membrane proteins via endocytosis, a process through which multiple ubiquitin ligases are known to specifically regulate the functions of a number of ion channels, transporters, and signaling receptors. Endocytic removal of these proteins through ubiquitin-dependent endocytosis provides a way to rapidly downregulate the physiological outcomes, and defects in such controls are directly linked to human pathologies. Recent evidence suggests that ubiquitination is also involved in the shedding of membranes and associated proteins as extracellular vesicles, thereby not only controlling the cell surface levels of some membrane proteins, but also their potential transport to neighboring cells. In this review, we summarize the mechanisms and functions of ubiquitination of membrane proteins and provide specific examples of ubiquitin-dependent regulation of membrane proteins.
Collapse
Affiliation(s)
- Natalie Foot
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Tanya Henshall
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| |
Collapse
|
42
|
Abstract
In the developing vertebrate embryo, segmentation initiates through the formation of repeated segments, or somites, on either side of the posterior neural tube along the anterior to posterior axis. The periodicity of somitogenesis is regulated by a molecular oscillator, the segmentation clock, driving cyclic gene expression in the unsegmented paraxial mesoderm, from which somites derive. Three signaling pathways underlie the molecular mechanism of the oscillator: Wnt, FGF, and Notch. In particular, Notch has been demonstrated to be an essential piece in the intricate somitogenesis regulation puzzle. Notch is required to synchronize oscillations between neighboring cells, and is moreover necessary for somite formation and clock gene oscillations. Following ligand activation, the Notch receptor is cleaved to liberate the active intracellular domain (NICD) and during somitogenesis NICD itself is produced and degraded in a cyclical manner, requiring tightly regulated, and coordinated turnover. It was recently shown that the pace of the segmentation clock is exquisitely sensitive to levels/stability of NICD. In this review, we focus on what is known about the mechanisms regulating NICD turnover, crucial to the activity of the pathway in all developmental contexts. To date, the regulation of NICD stability has been attributed to phosphorylation of the PEST domain which serves to recruit the SCF/Sel10/FBXW7 E3 ubiquitin ligase complex involved in NICD turnover. We will describe the pathophysiological relevance of NICD-FBXW7 interaction, whose defects have been linked to leukemia and a variety of solid cancers.
Collapse
Affiliation(s)
- Francesca A Carrieri
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee Dundee, UK
| | - Jacqueline Kim Dale
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee Dundee, UK
| |
Collapse
|
43
|
Callahan R, Chestnut BA, Raafat A. Original Research: Featured Article: Imatinib mesylate (Gleevec) inhibits Notch and c-Myc signaling: Five-day treatment permanently rescues mammary development. Exp Biol Med (Maywood) 2016; 242:53-67. [PMID: 27550925 DOI: 10.1177/1535370216665175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/28/2016] [Indexed: 12/31/2022] Open
Abstract
Wap-Int3 transgenic females expressing the Notch4 intracellular domain (designated Int3) from the whey acidic protein promoter exhibit two phenotypes in the mammary gland: blockage of lobuloalveolar development and lactation, and tumor development with 100% penetrance. Previously, we have shown that treatment of Wap-Int3 tumor bearing mice with Imatinib mesylate (Gleevec) is associated with complete regression of the tumor. In the present study, we show that treatment of Wap-Int3 mice during day 1 through day 6 of pregnancy with Gleevec leads to the restoration of their lobuloalveolar development and ability to lactate in subsequent pregnancies in absence of Gleevec treatment. In addition, these mice do not develop mammary tumors. We investigated the mechanism for Gleevec regulation of Notch signaling and found that Gleevec treatment results in a loss of Int3 protein but not of Int3 mRNA in HC11 mouse mammary epithelial cells expressing Int3. The addition of MG-132, a proteasome inhibitor, shows increased ubiquitination of Int3 in the presence of Gleevec. Thus, Gleevec affects the stability of Int3 by promoting the degradation of Int3 via E3 ubiquitin ligases targeting it for the proteasome degradation. Gleevec is a tyrosine kinase inhibitor that acts on c-Kit and PDGFR. Therefore, we investigated the downstream substrate kinase GSK3β to ascertain the possible role that this kinase might play in the stability of Int3. Data show that Gleevec degradation of Int3 is GSK3β dependent. We have expanded our study of the effects Gleevec has on tumorigenesis of other oncogenes. We have found that anchorage-independent growth of HC11-c-Myc cells as well as tumor growth in nude mice is inhibited by Gleevec treatment. As with Int3, Gleevec treatment appears to destabilize the c-Myc protein but not mRNA. These results indicate that Gleevec could be a potential therapeutic drug for patients bearing Notch4 and/or c-Myc positive breast carcinomas.
Collapse
Affiliation(s)
- Robert Callahan
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Barry A Chestnut
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ahmed Raafat
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
44
|
Liu J, Shen JX, Wen XF, Guo YX, Zhang GJ. Targeting Notch degradation system provides promise for breast cancer therapeutics. Crit Rev Oncol Hematol 2016; 104:21-9. [PMID: 27263934 DOI: 10.1016/j.critrevonc.2016.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 04/18/2016] [Accepted: 05/19/2016] [Indexed: 02/05/2023] Open
Abstract
Notch receptor signaling pathways play an important role, not only in normal breast development but also in breast cancer development and progression. As a group of ligand-induced proteins, different subtypes of mammalian Notch (Notch1-4) are sensitive to subtle changes in protein levels. Thus, a clear understanding of mechanisms of Notch protein turnover is essential for understanding normal and pathological mechanisms of Notch functions. It has been suggested that there is a close relationship between the carcinogenesis and the dysregulation of Notch degradation. However, this relationship remains mostly undefined in the context of breast cancer, as protein degradation is mediated by numerous signaling pathways as well as certain molecule modulators (activators/inhibitors). In this review, we summarize the published data regarding the regulation of Notch family member degradation in breast cancer, while emphasizing areas that are likely to provide new therapeutic modalities for mechanism-based anti-cancer drugs.
Collapse
Affiliation(s)
- Jing Liu
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Jia-Xin Shen
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Xiao-Fen Wen
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Yu-Xian Guo
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Guo-Jun Zhang
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| |
Collapse
|
45
|
Broadus MR, Chen TW, Neitzel LR, Ng VH, Jodoin JN, Lee LA, Salic A, Robbins DJ, Capobianco AJ, Patton JG, Huppert SS, Lee E. Identification of a Paralog-Specific Notch1 Intracellular Domain Degron. Cell Rep 2016; 15:1920-9. [PMID: 27210761 DOI: 10.1016/j.celrep.2016.04.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/14/2016] [Accepted: 04/19/2016] [Indexed: 01/13/2023] Open
Abstract
Upon Notch pathway activation, the receptor is cleaved to release the Notch intracellular domain (NICD), which translocates to the nucleus to activate gene transcription. Using Xenopus egg extracts, we have identified a Notch1-specific destruction signal (N1-Box). We show that mutations in the N1-Box inhibit NICD1 degradation and that the N1-Box is transferable for the promotion of degradation of heterologous proteins in Xenopus egg extracts and in cultured human cells. Mutation of the N1-Box enhances Notch1 activity in cultured human cells and zebrafish embryos. Human cancer mutations within the N1-Box enhance Notch1 signaling in transgenic zebrafish, highlighting the physiological relevance of this destruction signal. We find that binding of the Notch nuclear factor, CSL, to the N1-Box blocks NICD1 turnover. Our studies reveal a mechanism by which degradation of NICD1 is regulated by the N1-Box to minimize stochastic flux and to establish a threshold for Notch1 pathway activation.
Collapse
Affiliation(s)
- Matthew R Broadus
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Tony W Chen
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Leif R Neitzel
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Victoria H Ng
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeanne N Jodoin
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Laura A Lee
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - David J Robbins
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Anthony J Capobianco
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - James G Patton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Stacey S Huppert
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Ethan Lee
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
46
|
Regulation of Notch Signaling Through Intracellular Transport. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:107-27. [PMID: 26944620 DOI: 10.1016/bs.ircmb.2015.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The highly conserved Notch-signaling pathway performs a central role in cell differentiation, survival, and proliferation. A major mechanism by which cells modulate signaling is by controlling the intracellular transport itinerary of Notch. Indeed, Notch removal from the cell surface and its targeting to the lysosome for degradation is one way in which Notch activity is downregulated since it limits receptor exposure to ligand. In contrast, Notch-signaling capacity is maintained through repeated rounds of receptor recycling and redelivery of Notch to the cell surface from endosomal stores. This review discusses the molecular mechanisms by which Notch transit through the endosome is controlled and how various intracellular sorting decisions are thought to impact signaling activity.
Collapse
|
47
|
Mozūraitienė J, Bielskienė K, Atkočius V, Labeikytė D. Molecular alterations in signal pathways of melanoma and new personalized treatment strategies: Targeting of Notch. MEDICINA-LITHUANIA 2015; 51:133-145. [PMID: 28705475 DOI: 10.1016/j.medici.2015.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 04/14/2015] [Indexed: 02/09/2023]
Abstract
Despite modern achievements in therapy of malignant melanomas new treatment strategies are welcomed in clinics for survival of patients. Now it is supposed that personalized molecular therapies for each patient are needed concerning a specificity of molecular alterations in patient's tumors. In human melanoma, Notch signaling interacts with other pathways, including MAPK, PI3K-AKT, NF-kB, and p53. This article discusses mutated genes and leading aberrant signal pathways in human melanoma which are of interest concerning to their perspective for personalized treatment strategies in melanoma. We speculate that E3 ubiquitin ligases MDM2 and MDM4 can be attractive therapeutic target for p53 and Notch signaling pathways in malignant melanoma by using small molecule inhibitors. It is possible that restoration of p53-MDM2-NUMB complexes in melanoma can restore wild type p53 function and positively modulate Notch pathway. In this review we summarize recent data about novel US Food and Drug Administration approved target drugs for metastatic melanoma treatment, and suppose model for treatment strategy by targeting Notch.
Collapse
Affiliation(s)
| | - Kristina Bielskienė
- Department of Biochemistry and Molecular Biology, Vilnius University, Vilnius, Lithuania.
| | | | - Danutė Labeikytė
- Department of Biochemistry and Molecular Biology, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
48
|
Human Cytomegalovirus Infection Dysregulates the Localization and Stability of NICD1 and Jag1 in Neural Progenitor Cells. J Virol 2015; 89:6792-804. [PMID: 25903338 DOI: 10.1128/jvi.00351-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 04/10/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) infection of the developing fetus frequently results in major neural developmental damage. In previous studies, HCMV was shown to downregulate neural progenitor/stem cell (NPC) markers and induce abnormal differentiation. As Notch signaling plays a vital role in the maintenance of stem cell status and is a switch that governs NPC differentiation, the effect of HCMV infection on the Notch signaling pathway in NPCs was investigated. HCMV downregulated mRNA levels of Notch1 and its ligand, Jag1, and reduced protein levels and altered the intracellular localization of Jag1 and the intracellular effector form of Notch1, NICD1. These effects required HCMV gene expression and appeared to be mediated through enhanced proteasomal degradation. Transient expression of the viral tegument proteins of pp71 and UL26 reduced NICD1 and Jag1 protein levels endogenously and exogenously. Given the critical role of Notch signaling in NPC growth and differentiation, these findings reveal important mechanisms by which HCMV disturbs neural cell development in vitro. Similar events in vivo may be associated with HCMV-mediated neuropathogenesis during congenital infection in the fetal brain. IMPORTANCE Congenital human cytomegalovirus (HCMV) infection is the leading cause of birth defects that primarily manifest as neurological disabilities. Neural progenitor cells (NPCs), key players in fetal brain development, are the most susceptible cell type for HCMV infection in the fetal brain. Studies have shown that NPCs are fully permissive for HCMV infection, which causes neural cell loss and premature differentiation, thereby perturbing NPC fate. Elucidation of virus-host interactions that govern NPC proliferation and differentiation is critical to understanding neuropathogenesis. The Notch signaling pathway is critical for maintaining stem cell status and functions as a switch for differentiation of NPCs. Our investigation into the impact of HCMV infection on this pathway revealed that HCMV dysregulates Notch signaling by altering expression of the Notch ligand Jag1, Notch1, and its active effector in NPCs. These results suggest a mechanism for the neuropathogenesis induced by HCMV infection that includes altered NPC differentiation and proliferation.
Collapse
|
49
|
Bielskienė K, Bagdonienė L, Mozūraitienė J, Kazbarienė B, Janulionis E. E3 ubiquitin ligases as drug targets and prognostic biomarkers in melanoma. MEDICINA-LITHUANIA 2015; 51:1-9. [PMID: 25744769 DOI: 10.1016/j.medici.2015.01.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 01/18/2015] [Indexed: 12/30/2022]
Abstract
Melanomas are highly proliferative and invasive, and are most frequently metastatic. Despite many advances in cancer treatment over the last several decades, the prognosis for patients with advanced melanoma remains poor. New treatment methods and strategies are necessary. The main hallmark of cancer is uncontrolled cellular proliferation with alterations in the expression of proteins. Ubiquitin and ubiquitin-related proteins posttranslationally modify proteins and thereby alter their functions. The ubiquitination process is involved in various physiological responses, including cell growth, cell death, and DNA damage repair. E3 ligases, the most specific enzymes of ubiquitination system, participate in the turnover of many key regulatory proteins and in the development of cancer. E3 ligases are of interest as drug targets for their ability to regulate proteins stability and functions. Compared to the general proteasome inhibitor bortezomib, which blocks the entire protein degradation, drugs that target a particular E3 ligase are expected to have better selectivity with less associated toxicity. Components of different E3 ligases complexes (FBW7, MDM2, RBX1/ROC1, RBX2/ROC2, cullins and many others) are known as oncogenes or tumor suppressors in melanomagenesis. These proteins participate in regulation of different cellular pathways and such important proteins in cancer development as p53 and Notch. In this review we summarized published data on the role of known E3 ligases in the development of melanoma and discuss the inhibitors of E3 ligases as a novel approach for the treatment of malignant melanomas.
Collapse
Affiliation(s)
| | - Lida Bagdonienė
- Department of Biochemistry and Molecular Biology, Vilnius University, Vilnius, Lithuania.
| | | | | | | |
Collapse
|
50
|
Zhang Q, Zhang Y, Wu L, Yang Y, Li X, Gao L, Hou X, Wu Y, Hou G, Li Z, Lin X. dBrms1 Acts as a Positive Regulator of Notch Signaling in Drosophila Wing. J Genet Genomics 2014; 41:317-25. [DOI: 10.1016/j.jgg.2014.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/01/2014] [Accepted: 04/10/2014] [Indexed: 01/25/2023]
|