1
|
Miranda-Alban J, Sanchez-Luege N, Valbuena FM, Rangel C, Rebay I. The Abelson kinase and the Nedd4 family E3 ligases co-regulate Notch trafficking to limit signaling. J Cell Biol 2025; 224:e202407066. [PMID: 40183942 PMCID: PMC11970431 DOI: 10.1083/jcb.202407066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/25/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Precise output from the conserved Notch signaling pathway governs a plethora of cellular processes and developmental transitions. Unlike other pathways that use a cytoplasmic relay, the Notch cell surface receptor transduces signaling directly to the nucleus, with endocytic trafficking providing critical regulatory nodes. Here we report that the cytoplasmic tyrosine kinase Abelson (Abl) facilitates Notch internalization into late endosomes/multivesicular bodies (LEs), thereby limiting signaling output in both ligand-dependent and -independent contexts. Abl phosphorylates the PPxY motif within Notch, a molecular target for its degradation via Nedd4 family ubiquitin ligases. We show that Su(dx), a family member, mediates the Abl-directed LE regulation of Notch via the PPxY, while another family member, Nedd4Lo, contributes to Notch internalization into LEs through both PPxY-dependent and -independent mechanisms. Our findings demonstrate how a network of posttranslational modifiers converging at LEs cooperatively modulates Notch signaling to ensure the precision and robustness of its cellular and developmental functions.
Collapse
Affiliation(s)
- Julio Miranda-Alban
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL, USA
| | - Nicelio Sanchez-Luege
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL, USA
| | - Fernando M. Valbuena
- Cell and Molecular Biology Graduate Program, University of Chicago, Chicago, IL, USA
| | - Chyan Rangel
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Ilaria Rebay
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL, USA
- Cell and Molecular Biology Graduate Program, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Cheng Y, Sun Q, Chen Y, Wang J, Chen Y, Yang Y, Zhang J, Cao Y, Li Z, Zhang Y. DTX3 suppresses bladder cancer cell invasion and metastasis by inhibiting the Notch signaling pathway. Int Immunopharmacol 2025; 153:114529. [PMID: 40127622 DOI: 10.1016/j.intimp.2025.114529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025]
Abstract
Deltex E3 ubiquitin ligase 3 (DTX3) was identified as a tumor suppressor in human cancers. However, whether DTX3 could suppress the progression of bladder cancer (BC) remains unknown. In this study, DTX3 downregulation in BC tissues was confirmed at mRNA and protein levels, and decreased DTX3 expression was associated with poor prognosis. DTX3 knockdown triggered aberrant epithelial-to-mesenchymal transition (EMT), principally via downregulation of E-cadherin and upregulation of N-cadherin, MMP9, Snail, and Slug. Gain- and loss-of-function assays indicated that DTX3 acted as a suppressor gene by inhibiting the migration and invasion of BC cells both in vivo and in vitro. Further analysis revealed that DTX3 inhibited Notch signaling pathway activity, and the Notch signaling inhibitor DAPT could partially reverse the effects of DTX3 knockdown on the metastatic abilities of BC cells. Mechanically, DTX3 bind to Notch intracellular domain (NICD) via its C-terminal RING finger domain (RFD), ubiquitinated, and degraded NICD, resulting in repression of the Notch pathway. Our findings reveal the key role of DTX3 in binding to NICD, promoting its ubiquitination and protein degradation, and suppressing the activation of the Notch signaling pathway to inhibit BC invasion and metastasis.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pathology, Cancer Research Laboratory, Chengde Medical University, Chengde, China
| | - Qi Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ya Chen
- Department of Pathology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - JiaYu Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - YanJun Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - YuanZhong Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - JiangBo Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - ZhiYong Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - YiJun Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
3
|
Velikaneye BA, Kozak GM. Epigenomic Changes in Ostrinia Moths Under Elevated Pupal and Adult Temperature. Mol Ecol 2025; 34:e17676. [PMID: 39936612 DOI: 10.1111/mec.17676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
Epigenetic changes in the methylation of DNA may occur in response to environmental stressors, including warming climates. DNA methylation may also play an important role in regulating gene expression during both male and female reproduction in many insect species. However, it is currently unknown how DNA methylation shifts when individuals are reproducing under warmer temperatures. We exposed European corn borer moths (Ostrinia nubilalis) to heat during the pupal and adult life stages then investigated changes in DNA methylation across the genome using enzymatic methyl-seq (EM-seq). We compared methylation patterns in reproductive males and females exposed to heat (28°C) to those that experienced an ambient temperature (23°C). We found that heat exposure led to a small but significant increase in the percentage of methylated CpG sites throughout the genome in both sexes. However, DNA methylation rates were higher in females and differential methylation following heat exposure localised to unique regions in each sex. In males, methylation shifted within genes belonging to pathways including Hippo signalling, ubiquitin-mediated proteolysis, DNA damage repair and spermatogenesis. In females, differential methylation occurred in genes related to histone modification and oogenesis. Our results suggest that DNA methylation patterns respond to moderate heat exposure in Lepidoptera and provide insight into epigenetic responses to heatwaves, suggesting novel pathways that may be involved in responding to heat stress during metamorphosis and reproduction.
Collapse
Affiliation(s)
- Brittany A Velikaneye
- Department of Biology, University of Massachusetts Dartmouth, Dartmouth, Massachusetts, USA
| | - Genevieve M Kozak
- Department of Biology, University of Massachusetts Dartmouth, Dartmouth, Massachusetts, USA
| |
Collapse
|
4
|
Matusova Z, Dykstra W, de Pablo Y, Zetterdahl OG, Canals I, van Gelder CAGH, Vos HR, Pérez-Sala D, Kubista M, Abaffy P, Ahlenius H, Valihrach L, Hol EM, Pekny M. Aberrant neurodevelopment in human iPS cell-derived models of Alexander disease. Glia 2025; 73:57-79. [PMID: 39308436 DOI: 10.1002/glia.24618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 12/21/2024]
Abstract
Alexander disease (AxD) is a rare and severe neurodegenerative disorder caused by mutations in glial fibrillary acidic protein (GFAP). While the exact disease mechanism remains unknown, previous studies suggest that mutant GFAP influences many cellular processes, including cytoskeleton stability, mechanosensing, metabolism, and proteasome function. While most studies have primarily focused on GFAP-expressing astrocytes, GFAP is also expressed by radial glia and neural progenitor cells, prompting questions about the impact of GFAP mutations on central nervous system (CNS) development. In this study, we observed impaired differentiation of astrocytes and neurons in co-cultures of astrocytes and neurons, as well as in neural organoids, both generated from AxD patient-derived induced pluripotent stem (iPS) cells with a GFAPR239C mutation. Leveraging single-cell RNA sequencing (scRNA-seq), we identified distinct cell populations and transcriptomic differences between the mutant GFAP cultures and a corrected isogenic control. These findings were supported by results obtained with immunocytochemistry and proteomics. In co-cultures, the GFAPR239C mutation resulted in an increased abundance of immature cells, while in unguided neural organoids and cortical organoids, we observed altered lineage commitment and reduced abundance of astrocytes. Gene expression analysis revealed increased stress susceptibility, cytoskeletal abnormalities, and altered extracellular matrix and cell-cell communication patterns in the AxD cultures, which also exhibited higher cell death after stress. Overall, our results point to altered cell differentiation in AxD patient-derived iPS-cell models, opening new avenues for AxD research.
Collapse
Affiliation(s)
- Zuzana Matusova
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Werner Dykstra
- Department of Translational Neuroscience, University Medical Centre Utrecht Brain Centre, Utrecht University, Utrecht, The Netherlands
| | - Yolanda de Pablo
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Oskar G Zetterdahl
- Stem Cells, Aging and Neurodegeneration Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
- Glial and Neuronal Biology Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Isaac Canals
- Glial and Neuronal Biology Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Metabolism, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
- ITINERARE-Innovative therapies in rare diseases, University Research Priority Program, University of Zurich, Zurich, Switzerland
| | - Charlotte A G H van Gelder
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Harmjan R Vos
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Henrik Ahlenius
- Stem Cells, Aging and Neurodegeneration Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Centre Utrecht Brain Centre, Utrecht University, Utrecht, The Netherlands
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
5
|
O'Hare M, Miller WP, Arevalo-Alquichire S, Amarnani D, Apryani E, Perez-Corredor P, Marino C, Shu DY, Vanderleest TE, Muriel-Torres A, Gordon HB, Gunawan AL, Kaplan BA, Barake KW, Bejjani RP, Doan TH, Lin R, Delgado-Tirado S, Gonzalez-Buendia L, Rossin EJ, Zhao G, Eliott D, Weinl-Tenbruck C, Chevessier-Tünnesen F, Rejman J, Montrasio F, Kim LA, Arboleda-Velasquez JF. An mRNA-encoded dominant-negative inhibitor of transcription factor RUNX1 suppresses vitreoretinal disease in experimental models. Sci Transl Med 2024; 16:eadh0994. [PMID: 39602510 DOI: 10.1126/scitranslmed.adh0994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/06/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
Messenger RNA (mRNA)-based therapies are a promising approach to medical treatment. Except for infectious diseases, no other disease has mRNA-based therapies available. The eye is an ideal model for mRNA therapeutic development because it requires limited dosing. Proliferative vitreoretinopathy (PVR) is a blinding condition caused by retinal detachment that now lacks available medical treatment, with surgery as the only treatment option. We previously implicated runt-related transcription factor-1 (RUNX1) as a driver of epithelial-to-mesenchymal transition (EMT) in PVR and as a critical mediator of aberrant ocular angiogenesis when up-regulated. On the basis of these findings, an mRNA was designed to express a dominant-negative inhibitor of RUNX1 (RUNX1-Trap). We show that RUNX1-Trap delivered in polymer-lipidoid complexes or lipid nanoparticles sequestered RUNX1 in the cytosol and strongly reduced proliferation in primary cell cultures established from fibrotic membranes derived from patients with PVR. We assessed the preclinical efficacy of intraocular delivery of mRNA-encoded RUNX1-Trap in a rabbit model of PVR and in a laser-induced mouse model of aberrant angiogenesis often used to study wet age-related macular degeneration. mRNA-encoded RUNX1-Trap suppressed ocular pathology, measured as pathological scores in the rabbit PVR model and leakage and lesion size in the laser-induced choroidal neovascularization mouse model. mRNA-encoded RUNX1-Trap also strongly reduced proliferation in a human ex vivo explant model of PVR. These data demonstrate the therapeutic potential of mRNA-encoded therapeutic molecules with dominant-negative properties, highlighting the potential of mRNA-based therapies beyond standard gene supplementation approaches.
Collapse
Affiliation(s)
- Michael O'Hare
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - William P Miller
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Said Arevalo-Alquichire
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Dhanesh Amarnani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Evhy Apryani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Paula Perez-Corredor
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Claudia Marino
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Daisy Y Shu
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Timothy E Vanderleest
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Andres Muriel-Torres
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Harper B Gordon
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Audrey L Gunawan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Bryan A Kaplan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Karim W Barake
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Romy P Bejjani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Tri H Doan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Rose Lin
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Santiago Delgado-Tirado
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Lucia Gonzalez-Buendia
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Elizabeth J Rossin
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Guannan Zhao
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Dean Eliott
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | - Leo A Kim
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
6
|
Ramsey KM, Barrick D. One RING to rule them all: Mind bomb-1 ccRING3 controls Notch signaling. Structure 2024; 32:1550-1551. [PMID: 39366337 DOI: 10.1016/j.str.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
In this issue of Structure, Cao et al.1 use X-ray crystallography, biochemical, and genetic studies to define the key role of the Mind bomb-1 ccRING3 domain in triggering Notch signaling, and they demonstrate that ccRING3-mediated dimerization is a key step in ligand activation.
Collapse
Affiliation(s)
- Kristen M Ramsey
- T.C. Jenkins Department of Biophysics, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21209, USA.
| | - Doug Barrick
- T.C. Jenkins Department of Biophysics, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21209, USA
| |
Collapse
|
7
|
Sachan N, Sharma V, Mutsuddi M, Mukherjee A. Notch signalling: multifaceted role in development and disease. FEBS J 2024; 291:3030-3059. [PMID: 37166442 DOI: 10.1111/febs.16815] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/08/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Notch pathway is an evolutionarily conserved signalling system that operates to influence an astonishing array of cell fate decisions in different developmental contexts. Notch signalling plays important roles in many developmental processes, making it difficult to name a tissue or a developing organ that does not depend on Notch function at one stage or another. Thus, dysregulation of Notch signalling is associated with many developmental defects and various pathological conditions, including cancer. Although many recent advances have been made to reveal different aspects of the Notch signalling mechanism and its intricate regulation, there are still many unanswered questions related to how the Notch signalling pathway functions in so many developmental events. The same pathway can be deployed in numerous cellular contexts to play varied and critical roles in an organism's development and this is only possible because of the complex regulatory mechanisms of the pathway. In this review, we provide an overview of the mechanism and regulation of the Notch signalling pathway along with its multifaceted functions in different aspects of development and disease.
Collapse
Affiliation(s)
- Nalani Sachan
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - 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
|
8
|
Wilkin MB, Whiteford R, Akbar T, Hosseini-Alghaderi S, Revici R, Carbery AM, Baron M. The First Defined Null Allele of the Notch Regulator, a Suppressor of Deltex: Uncovering Its Novel Roles in Drosophila melanogaster Oogenesis. Biomolecules 2024; 14:522. [PMID: 38785929 PMCID: PMC11118177 DOI: 10.3390/biom14050522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Suppressor of deltex (Su(dx)) is a Drosophila melanogaster member of the NEDD4 family of the HECT domain E3 ubiquitin ligases. Su(dx) acts as a regulator of Notch endocytic trafficking, promoting Notch lysosomal degradation and the down-regulation of both ligand-dependent and ligand-independent signalling, the latter involving trafficking through the endocytic pathway and activation of the endo/lysosomal membrane. Mutations of Su(dx) result in developmental phenotypes in the Drosophila wing that reflect increased Notch signalling, leading to gaps in the specification of the wing veins, and Su(dx) functions to provide the developmental robustness of Notch activity to environmental temperature shifts. The full developmental functions of Su(dx) are unclear; however, this is due to a lack of a clearly defined null allele. Here we report the first defined null mutation of Su(dx), generated by P-element excision, which removes the complete open reading frame. We show that the mutation is recessive-viable, with the Notch gain of function phenotypes affecting wing vein and leg development. We further uncover new roles for Su(dx) in Drosophila oogenesis, where it regulates interfollicular stalk formation, egg chamber separation and germline cyst enwrapment by the follicle stem cells. Interestingly, while the null allele exhibited a gain in Notch activity during oogenesis, the previously described Su(dx)SP allele, which carries a seven amino acid in-frame deletion, displayed a Notch loss of function phenotypes and an increase in follicle stem cell turnover. This is despite both alleles displaying similar Notch gain of function in wing development. We attribute this unexpected context-dependent outcome of Su(dx)sp being due to the partial retention of function by the intact C2 and WW domain regions of the protein. Our results extend our understanding of the developmental role of Su(dx) in the tissue renewal and homeostasis of the Drosophila ovary and illustrate the importance of examining an allelic series of mutations to fully understand developmental functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Martin Baron
- Manchester Academic Health Science Centre, School of Biological Sciences, University of Manchester, Michael Smith Building and Oxford Rd., Manchester M13 9PT, UK
| |
Collapse
|
9
|
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
|
10
|
Kaur D, Khan H, Grewal AK, Singh TG. Glycosylation: A new signaling paradigm for the neurovascular diseases. Life Sci 2024; 336:122303. [PMID: 38016576 DOI: 10.1016/j.lfs.2023.122303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023]
Abstract
A wide range of life-threatening conditions with complicated pathogenesis involves neurovascular disorders encompassing Neurovascular unit (NVU) damage. The pathophysiology of NVU is characterized by several features including tissue hypoxia, stimulation of inflammatory and angiogenic processes, and the initiation of intricate molecular interactions, collectively leading to an elevation in blood-brain barrier permeability, atherosclerosis and ultimately, neurovascular diseases. The presence of compelling data about the significant involvement of the glycosylation in the development of diseases has sparked a discussion on whether the abnormal glycosylation may serve as a causal factor for neurovascular disorders, rather than being just recruited as a secondary player in regulating the critical events during the development processes like embryo growth and angiogenesis. An essential tool for both developing new anti-ischemic therapies and understanding the processes of ischemic brain damage is undertaking pre-clinical studies of neurovascular disorders. Together with the post-translational modification of proteins, the modulation of glycosylation and its enzymes implicates itself in several abnormal activities which are known to accelerate neuronal vasculopathy. Despite the failure of the majority of glycosylation-based preclinical and clinical studies over the past years, there is a significant probability to provide neuroprotection utilizing modern and advanced approaches to target abnormal glycosylation activity at embryonic stages as well. This article focuses on a variety of experimental evidence to postulate the interconnection between glycosylation and vascular disorders along with possible treatment options.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | | | | |
Collapse
|
11
|
Cai J, Qiao Y, Chen L, Lu Y, Zheng D. Regulation of the Notch signaling pathway by natural products for cancer therapy. J Nutr Biochem 2024; 123:109483. [PMID: 37848105 DOI: 10.1016/j.jnutbio.2023.109483] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
The Notch signaling pathway is an evolutionarily conserved pathway that modulates normal biological processes involved in cellular differentiation, apoptosis, and stem cell self-renewal in a context-dependent fashion. Attributed to its pleiotropic physiological roles, both overexpression and silencing of the pathway are associated with the emergence, progression, and poorer prognosis in various types of cancer. To decrease disease incidence and promote survival, targeting Notch may have chemopreventive and anti-cancer effects. Natural products with profound historical origins have distinguished themselves from other therapies due to their easy access, high biological compatibility, low toxicity, and reliable effects at specific physiological sites in vivo. This review describes the Notch signaling pathway, particularly its normal activation process, and some main illnesses related to Notch signaling pathway dysregulation. Emphasis is placed on the effects and mechanisms of natural products targeting the Notch signaling pathway in diverse cancer types, including curcumin, ellagic acid (EA), resveratrol, genistein, epigallocatechin-3-gallate (EGCG), quercetin, and xanthohumol and so on. Existing evidence indicates that natural products are feasible solution to fight against cancer by targeting Notch signaling, either alone or in combination with current therapeutic agents.
Collapse
Affiliation(s)
- Jiayi Cai
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Yajie Qiao
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Lingbin Chen
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Youguang Lu
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China; Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350001, China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
12
|
Ma Z, Zeng Y, Wang M, Liu W, Zhou J, Wu C, Hou L, Yin B, Qiang B, Shu P, Peng X. N4BP1 mediates RAM domain-dependent notch signaling turnover during neocortical development. EMBO J 2023; 42:e113383. [PMID: 37807845 PMCID: PMC10646556 DOI: 10.15252/embj.2022113383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
Notch signaling pathway activity, particularly fluctuations in the biologically active effector fragment NICD, is required for rapid and efficient dynamic regulation of proper fate decisions in stem cells. In this study, we identified NEDD4-binding protein 1 (N4BP1), which is highly expressed in the developing mouse cerebral cortex, as a negative modulator of Notch signaling dynamics in neural progenitor cells. Intriguingly, N4BP1 regulated NICD stability specifically after Notch1 S3 cleavage through ubiquitin-mediated degradation that depended on its RAM domain, not its PEST domain, as had been extensively and previously described. The CoCUN domain in N4BP1, particularly the "Phe-Pro" motif (862/863 amino acid), was indispensable for mediating NICD degradation. The Ring family E3 ligase Trim21 was, in contrast to other NEDD4 family members, required for N4BP1-regulated NICD degradation. Overexpression of N4BP1 in cortical neural progenitors promoted neural stem cell differentiation, whereas neural progenitor cells lacking N4BP1 were sensitized to Notch signaling, resulting in the maintenance of stem-like properties in neural progenitor cells and lower production of cortical neurons.
Collapse
Affiliation(s)
- Zhihua Ma
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Yi Zeng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Present address:
Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)The Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ming Wang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Present address:
Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren HospitalCapital Medical University, Beijing Key Laboratory of Nasal Diseases, Beijing Institute of OtolaryngologyBeijingChina
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Jiafeng Zhou
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Chao Wu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Lin Hou
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Bin Yin
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Boqin Qiang
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major DiseasesBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| | - Xiaozhong Peng
- Department of Molecular Biology and Biochemistry, Medical Primate Research Center, Neuroscience CenterInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Respiratory Health and MultimorbidityBeijingChina
- Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| |
Collapse
|
13
|
Datkhayev UM, Rakhmetova V, Shepetov AM, Kodasbayev A, Datkayeva GM, Pazilov SB, Farooqi AA. Unraveling the Complex Web of Mechanistic Regulation of Versatile NEDD4 Family by Non-Coding RNAs in Carcinogenesis and Metastasis: From Cell Culture Studies to Animal Models. Cancers (Basel) 2023; 15:3971. [PMID: 37568787 PMCID: PMC10417118 DOI: 10.3390/cancers15153971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 08/13/2023] Open
Abstract
Discoveries related to an intriguing feature of ubiquitination have prompted a detailed analysis of the ubiquitination patterns in malignant cells. How the "ubiquitinome" is reshaped during multistage carcinogenesis has garnered significant attention. Seminal studies related to the structural and functional characterization of NEDD4 (Neuronal precursor cell-expressed developmentally downregulated-4) have consolidated our understanding at a new level of maturity. Additionally, regulatory roles of non-coding RNAs have further complicated the complex interplay between non-coding RNAs and the members of NEDD4 family. These mechanisms range from the miRNA-mediated targeting of NEDD4 family members to the regulation of transcriptional factors for a broader range of non-coding RNAs. Additionally, the NEDD4-mediated degradation of different proteins is modulated by lncRNAs and circRNAs. The miRNA-mediated targeting of NEDD4 family members is also regulated by circRNAs. Tremendous advancements have been made in the identification of different substrates of NEDD4 family and in the comprehensive analysis of the molecular mechanisms by which various members of NEDD4 family catalyze the ubiquitination of substrates. In this review, we have attempted to summarize the multifunctional roles of the NEDD4 family in cancer biology, and how different non-coding RNAs modulate these NEDD4 family members in the regulation of cancer. Future molecular studies should focus on the investigation of a broader drug design space and expand the scope of accessible targets for the inhibition/prevention of metastasis.
Collapse
Affiliation(s)
- Ubaidilla M. Datkhayev
- Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan
| | | | - Abay M. Shepetov
- Department of Nephrology, Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan;
| | - Almat Kodasbayev
- Department of Cardiovascular Surgery, Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan
| | | | - Sabit B. Pazilov
- Department of Healthcare of Kyzylorda Region, Kyzylorda, Abay Avenue, 27, Kyzylorda 120008, Kazakhstan;
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan
| |
Collapse
|
14
|
Toribio ML, González-García S. Notch Partners in the Long Journey of T-ALL Pathogenesis. Int J Mol Sci 2023; 24:1383. [PMID: 36674902 PMCID: PMC9866461 DOI: 10.3390/ijms24021383] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological disease that arises from the oncogenic transformation of developing T cells during T-lymphopoiesis. Although T-ALL prognosis has improved markedly in recent years, relapsing and refractory patients with dismal outcomes still represent a major clinical issue. Consequently, understanding the pathological mechanisms that lead to the appearance of this malignancy and developing novel and more effective targeted therapies is an urgent need. Since the discovery in 2004 that a major proportion of T-ALL patients carry activating mutations that turn NOTCH1 into an oncogene, great efforts have been made to decipher the mechanisms underlying constitutive NOTCH1 activation, with the aim of understanding how NOTCH1 dysregulation converts the physiological NOTCH1-dependent T-cell developmental program into a pathological T-cell transformation process. Several molecular players have so far been shown to cooperate with NOTCH1 in this oncogenic process, and different therapeutic strategies have been developed to specifically target NOTCH1-dependent T-ALLs. Here, we comprehensively analyze the molecular bases of the cross-talk between NOTCH1 and cooperating partners critically involved in the generation and/or maintenance and progression of T-ALL and discuss novel opportunities and therapeutic approaches that current knowledge may open for future treatment of T-ALL patients.
Collapse
Affiliation(s)
- María Luisa Toribio
- Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | | |
Collapse
|
15
|
Yousefi H, Bahramy A, Zafari N, Delavar MR, Nguyen K, Haghi A, Kandelouei T, Vittori C, Jazireian P, Maleki S, Imani D, Moshksar A, Bitaraf A, Babashah S. Notch signaling pathway: a comprehensive prognostic and gene expression profile analysis in breast cancer. BMC Cancer 2022; 22:1282. [PMID: 36476410 PMCID: PMC9730604 DOI: 10.1186/s12885-022-10383-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a complex disease exhibiting a great degree of heterogeneity due to different molecular subtypes. Notch signaling regulates the differentiation of breast epithelial cells during normal development and plays a crucial role in breast cancer progression through the abnormal expression of the Notch up-and down-stream effectors. To date, there are only a few patient-centered clinical studies using datasets characterizing the role of Notch signaling pathway regulators in breast cancer; thus, we investigate the role and functionality of these factors in different subtypes using publicly available databases containing records from large studies. High-throughput genomic data and clinical information extracted from TCGA were analyzed. We performed Kaplan-Meier survival and differential gene expression analyses using the HALLMARK_NOTCH_SIGNALING gene set. To determine if epigenetic regulation of the Notch regulators contributes to their expression, we analyzed methylation levels of these factors using the TCGA HumanMethylation450 Array data. Notch receptors and ligands expression is generally associated with the tumor subtype, grade, and stage. Furthermore, we showed gene expression levels of most Notch factors were associated with DNA methylation rate. Modulating the expression levels of Notch receptors and effectors can be a potential therapeutic approach for breast cancer. As we outline herein, elucidating the novel prognostic and regulatory roles of Notch implicate this pathway as an essential mediator controlling breast cancer progression.
Collapse
Affiliation(s)
- Hassan Yousefi
- Biochemistry & Molecular Biology, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, USA
| | - Afshin Bahramy
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Zafari
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Khoa Nguyen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Atousa Haghi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Kandelouei
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Cecilia Vittori
- Louisiana State University Health Sciences Center (LSUHSC), and Stanley S. Scott Cancer Center, New Orleans, LA, USA
| | - Parham Jazireian
- Department of Biology, University Campus 2, University of Guilan, Rasht, Iran
| | - Sajad Maleki
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Danyal Imani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amin Moshksar
- Interventional Radiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran.
| |
Collapse
|
16
|
Tamaki Y, Shibata Y, Hayakawa M, Kato N, Machii A, Ikeda Y, Nanizawa E, Hayashi Y, Suemizu H, Ito H, Ishikawa T. Treatment with hepatocyte transplantation in a novel mouse model of persistent liver failure. Biochem Biophys Rep 2022; 32:101382. [DOI: 10.1016/j.bbrep.2022.101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
|
17
|
Begum F, Keni R, Ahuja TN, Beegum F, Nandakumar K, Shenoy RR. Notch signaling: A possible therapeutic target and its role in diabetic foot ulcers. Diabetes Metab Syndr 2022; 16:102542. [PMID: 35724488 DOI: 10.1016/j.dsx.2022.102542] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND & AIM Diabetic foot ulcers are major cause of lower limb amputations in the diabetic population. The major factors that play a role in causing the delay of the process of healing in diabetic foot ulcers broadly are decreased angiogenesis, reduced proliferation and migration of keratinocytes/fibroblasts. The typical wound healing process has four phases which are overlapping with each other thus making the healing even more complex. Hence it is essential to identify a therapeutic target that involves the regulation of the cellular factors involved in healing and helps to increase angiogenesis and can regulate all four phases accordingly. METHOD Literature review involved a search of the databases namely, PubMed, Cochrane, EMBASE, and Web of Science database. Articles were identified and retrieved that specifically dealt with Notch as a target in healing of wounds and its mechanism of action on various cells and phases of healing. RESULTS Notch is a cell surface receptor which interacts with transmembrane ligands of the nearby cells and is involved in cell proliferation, differentiation, cell fate and death. It is also involved in cell-to-cell communication, cell signaling, and various phases of development. There exist four known notch genes and five ligands which interact with notch proteins. Hyperglycemia plays a role in the activation of the notch receptor thus causing the release of inflammatory mediators via macrophages. As notch can regulate macrophage-mediated inflammation it can serve as a therapeutic target for diabetic foot ulcers. CONCLUSION This review focuses on the effect of notch on various cell mediators and phases of diabetic wound healing and deals with how notch activation or inhibition can serve as a potential therapeutic target for healing diabetic foot ulcers.
Collapse
Affiliation(s)
- Farmiza Begum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raghuvir Keni
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Tejas N Ahuja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Rekha R Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
18
|
Dean DM, Deitcher DL, Paster CO, Xu M, Loehlin DW. "A fly appeared": sable, a classic Drosophila mutation, maps to Yippee, a gene affecting body color, wings, and bristles. G3 (BETHESDA, MD.) 2022; 12:jkac058. [PMID: 35266526 PMCID: PMC9073688 DOI: 10.1093/g3journal/jkac058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/04/2022] [Indexed: 11/12/2022]
Abstract
Insect body color is an easily assessed and visually engaging trait that is informative on a broad range of topics including speciation, biomaterial science, and ecdysis. Mutants of the fruit fly Drosophila melanogaster have been an integral part of body color research for more than a century. As a result of this long tenure, backlogs of body color mutations have remained unmapped to their genes, all while their strains have been dutifully maintained, used for recombination mapping, and part of genetics education. Stemming from a lesson plan in our undergraduate genetics class, we have mapped sable1, a dark body mutation originally described by Morgan and Bridges, to Yippee, a gene encoding a predicted member of the E3 ubiquitin ligase complex. Deficiency/duplication mapping, genetic rescue, DNA and cDNA sequencing, RT-qPCR, and 2 new CRISPR alleles indicated that sable1 is a hypomorphic Yippee mutation due to an mdg4 element insertion in the Yippee 5'-UTR. Further analysis revealed additional Yippee mutant phenotypes including curved wings, ectopic/missing bristles, delayed development, and failed adult emergence. RNAi of Yippee in the ectoderm phenocopied sable body color and most other Yippee phenotypes. Although Yippee remains functionally uncharacterized, the results presented here suggest possible connections between melanin biosynthesis, copper homeostasis, and Notch/Delta signaling; in addition, they provide insight into past studies of sable cell nonautonomy and of the genetic modifier suppressor of sable.
Collapse
Affiliation(s)
- Derek M Dean
- Department of Biology, Williams College, Williamstown, MA 01267, USA
| | - David L Deitcher
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14853, USA
| | - Caleigh O Paster
- Department of Biology, Williams College, Williamstown, MA 01267, USA
| | - Manting Xu
- Department of Biology, Williams College, Williamstown, MA 01267, USA
| | - David W Loehlin
- Department of Biology, Williams College, Williamstown, MA 01267, USA
| |
Collapse
|
19
|
Okubo Y, Ohtake F, Igarashi K, Yasuhiko Y, Hirabayashi Y, Saga Y, Kanno J. Cleaved Delta like 1 intracellular domain regulates neural development via Notch signal-dependent and -independent pathways. Development 2021; 148:272156. [PMID: 34519339 PMCID: PMC8513606 DOI: 10.1242/dev.193664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/06/2021] [Indexed: 11/20/2022]
Abstract
Notch-Delta signaling regulates many developmental processes, including tissue homeostasis and maintenance of stem cells. Upon interaction of juxtaposed cells via Notch and Delta proteins, intracellular domains of both transmembrane proteins are cleaved and translocate to the nucleus. Notch intracellular domain activates target gene expression; however, the role of the Delta intracellular domain remains elusive. Here, we show the biological function of Delta like 1 intracellular domain (D1ICD) by modulating its production. We find that the sustained production of D1ICD abrogates cell proliferation but enhances neurogenesis in the developing dorsal root ganglia (DRG), whereas inhibition of D1ICD production promotes cell proliferation and gliogenesis. D1ICD acts as an integral component of lateral inhibition mechanism by inhibiting Notch activity. In addition, D1ICD promotes neurogenesis in a Notch signaling-independent manner. We show that D1ICD binds to Erk1/2 in neural crest stem cells and inhibits the phosphorylation of Erk1/2. In summary, our results indicate that D1ICD regulates DRG development by modulating not only Notch signaling but also the MAP kinase pathway.
Collapse
Affiliation(s)
- Yusuke Okubo
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Fumiaki Ohtake
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan.,Institute for Advanced Life Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Katsuhide Igarashi
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan.,Life Science Tokyo Advanced Research center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Science, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yukuto Yasuhiko
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Yoko Hirabayashi
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Yumiko Saga
- Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima 411-8540, Japan.,Department of Biological Science, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jun Kanno
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| |
Collapse
|
20
|
Xiu M, Zeng X, Shan R, Wen W, Li J, Wan R. Targeting Notch4 in Cancer: Molecular Mechanisms and Therapeutic Perspectives. Cancer Manag Res 2021; 13:7033-7045. [PMID: 34526819 PMCID: PMC8436177 DOI: 10.2147/cmar.s315511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/03/2021] [Indexed: 12/28/2022] Open
Abstract
The dysregulation of Notch signaling is found in many cancers and is closely related to cancer progression. As an important Notch receptor, abnormal Notch4 expression affects several tumor-cell behaviors, including stemness, the epithelial-mesenchymal transition, radio/chemoresistance and angiogenesis. In order to inhibit the oncogenic effects of Notch4 activation, several methods for targeting Notch4 signaling have been proposed. In this review, we summarize the known molecular mechanisms through which Notch4 affects cancer progression. Finally, we discuss potential Notch4-targeting therapeutic strategies as a reference for future research.
Collapse
Affiliation(s)
- Mengxi Xiu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China.,Second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Xiaohong Zeng
- Imaging Department, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Renfeng Shan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Wu Wen
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Jianfeng Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| | - Renhua Wan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
21
|
Revisiting Ehrlichia ruminantium Replication Cycle Using Proteomics: The Host and the Bacterium Perspectives. Microorganisms 2021; 9:microorganisms9061144. [PMID: 34073568 PMCID: PMC8229282 DOI: 10.3390/microorganisms9061144] [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: 01/15/2021] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
The Rickettsiales Ehrlichia ruminantium, the causal agent of the fatal tick-borne disease Heartwater, induces severe damage to the vascular endothelium in ruminants. Nevertheless, E. ruminantium-induced pathobiology remains largely unknown. Our work paves the way for understanding this phenomenon by using quantitative proteomic analyses (2D-DIGE-MS/MS, 1DE-nanoLC-MS/MS and biotin-nanoUPLC-MS/MS) of host bovine aorta endothelial cells (BAE) during the in vitro bacterium intracellular replication cycle. We detect 265 bacterial proteins (including virulence factors), at all time-points of the E. ruminantium replication cycle, highlighting a dynamic bacterium–host interaction. We show that E. ruminantium infection modulates the expression of 433 host proteins: 98 being over-expressed, 161 under-expressed, 140 detected only in infected BAE cells and 34 exclusively detected in non-infected cells. Cystoscape integrated data analysis shows that these proteins lead to major changes in host cell immune responses, host cell metabolism and vesicle trafficking, with a clear involvement of inflammation-related proteins in this process. Our findings led to the first model of E. ruminantium infection in host cells in vitro, and we highlight potential biomarkers of E. ruminantium infection in endothelial cells (such as ROCK1, TMEM16K, Albumin and PTPN1), which may be important to further combat Heartwater, namely by developing non-antibiotic-based strategies.
Collapse
|