51
|
Abstract
Vascularization is a major hurdle in complex tissue and organ engineering. Tissues greater than 200 μm in diameter cannot rely on simple diffusion to obtain nutrients and remove waste. Therefore, an integrated vascular network is required for clinical translation of engineered tissues. Microvessels have been described as <150 μm in diameter, but clinically they are defined as <1 mm. With new advances in super microsurgery, vessels less than 1 mm can be anastomosed to the recipient circulation. However, this technical advancement still relies on the creation of a stable engineered microcirculation that is amenable to surgical manipulation and is readily perfusable. Microvascular engineering lays on the crossroads of microfabrication, microfluidics, and tissue engineering strategies that utilize various cellular constituents. Early research focused on vascularization by co-culture and cellular interactions, with the addition of angiogenic growth factors to promote vascular growth. Since then, multiple strategies have been utilized taking advantage of innovations in additive manufacturing, biomaterials, and cell biology. However, the anatomy and dynamics of native blood vessels has not been consistently replicated. Inconsistent results can be partially attributed to cell sourcing which remains an enigma for microvascular engineering. Variations of endothelial cells, endothelial progenitor cells, and stem cells have all been used for microvascular network fabrication along with various mural cells. As each source offers advantages and disadvantages, there continues to be a lack of consensus. Furthermore, discord may be attributed to incomplete understanding about cell isolation and characterization without considering the microvascular architecture of the desired tissue/organ.
Collapse
|
52
|
Tasdemir N, Ding K, Savariau L, Levine KM, Du T, Elangovan A, Bossart EA, Lee AV, Davidson NE, Oesterreich S. Proteomic and transcriptomic profiling identifies mediators of anchorage-independent growth and roles of inhibitor of differentiation proteins in invasive lobular carcinoma. Sci Rep 2020; 10:11487. [PMID: 32661241 PMCID: PMC7359337 DOI: 10.1038/s41598-020-68141-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
Invasive lobular carcinoma (ILC) is a histological subtype of breast cancer with distinct molecular and clinical features from the more common subtype invasive ductal carcinoma (IDC). ILC cells exhibit anchorage-independent growth in ultra-low attachment (ULA) suspension cultures, which is largely attributed to the loss of E-cadherin. In addition to anoikis resistance, herein we show that human ILC cell lines exhibit enhanced cell proliferation in ULA cultures as compared to IDC cells. Proteomic comparison of ILC and IDC cell lines identified induction of PI3K/Akt and p90-RSK pathways specifically in ULA culture in ILC cells. Further transcriptional profiling uncovered unique upregulation of the inhibitors of differentiation family transcription factors ID1 and ID3 in ILC ULA culture, the knockdown of which diminished the anchorage-independent growth of ILC cell lines through cell cycle arrest. We find that ID1 and ID3 expression is higher in human ILC tumors as compared to IDC, correlated with worse prognosis uniquely in patients with ILC and associated with upregulation of angiogenesis and matrisome-related genes. Altogether, our comprehensive study of anchorage independence in human ILC cell lines provides mechanistic insights and clinical implications for metastatic dissemination of ILC and implicates ID1 and ID3 as novel drivers and therapeutic targets for lobular breast cancer.
Collapse
Affiliation(s)
- Nilgun Tasdemir
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Kai Ding
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Laura Savariau
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, 15261, USA
| | - Kevin M Levine
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Tian Du
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ashuvinee Elangovan
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Emily A Bossart
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Adrian V Lee
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- University of Washington, Seattle, WA, 98195, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA, 15213, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
53
|
Wang X, Zhao Y, Fei X, Lu Q, Li Y, Yuan Y, Lu C, Li C, Chen H. LEF1/Id3/HRAS axis promotes the tumorigenesis and progression of esophageal squamous cell carcinoma. Int J Biol Sci 2020; 16:2392-2404. [PMID: 32760207 PMCID: PMC7378645 DOI: 10.7150/ijbs.47035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/10/2020] [Indexed: 01/04/2023] Open
Abstract
Our previous study demonstrated that lymphoid enhancer-binding factor 1 (LEF1) could promote the progression of esophageal squamous cell carcinoma (ESCC). However, the regulatory mechanism of LEF1 was not clear thoroughly. Herein, we continued to explore the downstream mechanism of LEF1 in ESCC. In this study, we applied western blotting, quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry, RNA-Seq analysis, a luciferase reporter assay, chromatin immunoprecipitation (ChIP), bioinformatics analysis, and a series of functional assays in vitro and in vivo. The results demonstrated that LEF1 regulated directly the expression of Id3. Id3 was highly expressed in ESCC tissues and correlated with histologic differentiation (p=0.011), pT stage (p<0.01) and AJCC stage (p<0.01) in ESCC patients. Moreover, Id3 could serve as a prognostic factor of ESCC. By various functional experiments, overexpression of Id3 promoted the proliferation, migration, invasion, EMT, and tumorgenicity. Mechanistically, Id3 could regulate ERK/MAPK signaling pathway via activating HRAS to perform its biological function. Furthermore, activating ERK/MAPK signaling pathway promoted the expression of Id3 gene in turn, indicating that a positive regulatory loop between Id3 and ERK/MAPK pathway may exist in ESCC. In summary, LEF1/Id3/HRAS axis could promote the tumorigenesis and progression of ESCC via activating ERK/MAPK signaling pathway. Targeting this cascade may provide a valid antitumor strategy to delay ESCC progress.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yue Zhao
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xiang Fei
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Qijue Lu
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yang Li
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yang Yuan
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chaojing Lu
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chunguang Li
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Hezhong Chen
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
54
|
Lee MY, Lee J, Hyeon SJ, Cho H, Hwang YJ, Shin J, McKee AC, Kowall NW, Kim J, Stein TD, Hwang D, Ryu H. Epigenome signatures landscaped by histone H3K9me3 are associated with the synaptic dysfunction in Alzheimer's disease. Aging Cell 2020; 19:e13153. [PMID: 32419307 PMCID: PMC7294781 DOI: 10.1111/acel.13153] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 02/09/2020] [Accepted: 03/29/2020] [Indexed: 01/03/2023] Open
Abstract
The pathogenesis of Alzheimer's disease (AD) and the commonest cause of dementia in the elderly remain incompletely understood. Recently, epigenetic modifications have been shown to play a potential role in neurodegeneration, but the specific involvement of epigenetic signatures landscaped by heterochromatin has not been studied in AD. Herein, we discovered that H3K9me3-mediated heterochromatin condensation is elevated in the cortex of sporadic AD postmortem brains. In order to identify which epigenomes are modulated by heterochromatin, we performed H3K9me3-chromatin immunoprecipitation (ChIP)-sequencing and mRNA-sequencing on postmortem brains from normal subjects and AD patients. The integrated analyses of genome-wide ChIP- and mRNA-sequencing data identified epigenomes that were highly occupied by H3K9me3 and inversely correlated with their mRNA expression levels in AD. Biological network analysis further revealed H3K9me3-landscaped epigenomes to be mainly involved in synaptic transmission, neuronal differentiation, and cell motility. Together, our data show that the abnormal heterochromatin remodeling by H3K9me3 leads to down-regulation of synaptic function-related genes, suggesting that the epigenetic alteration by H3K9me3 is associated with the synaptic pathology of sporadic AD.
Collapse
Affiliation(s)
| | - Junghee Lee
- Veteran's Affairs Boston Healthcare SystemBostonMAUSA,Department of NeurologyBoston University Alzheimer’s Disease CenterBoston University School of MedicineBostonMAUSA
| | - Seung Jae Hyeon
- Center for NeuromedicineBrain Science InstituteKorea Institute of Science and TechnologySeoulSouth Korea
| | - Hyesun Cho
- Genome Medicine Institute and Department of BiochemistrySeoul National University College of MedicineSeoulSouth Korea
| | - Yu Jin Hwang
- Center for NeuromedicineBrain Science InstituteKorea Institute of Science and TechnologySeoulSouth Korea
| | - Jong‐Yeon Shin
- Genome Medicine Institute and Department of BiochemistrySeoul National University College of MedicineSeoulSouth Korea
| | - Ann C. McKee
- Veteran's Affairs Boston Healthcare SystemBostonMAUSA,Department of NeurologyBoston University Alzheimer’s Disease CenterBoston University School of MedicineBostonMAUSA,Center for the Study of Traumatic EncephalopathyBoston University School of MedicineBostonMAUSA
| | - Neil W. Kowall
- Veteran's Affairs Boston Healthcare SystemBostonMAUSA,Department of NeurologyBoston University Alzheimer’s Disease CenterBoston University School of MedicineBostonMAUSA
| | - Jong‐Il Kim
- Genome Medicine Institute and Department of BiochemistrySeoul National University College of MedicineSeoulSouth Korea
| | - Thor D. Stein
- Veteran's Affairs Boston Healthcare SystemBostonMAUSA,Department of NeurologyBoston University Alzheimer’s Disease CenterBoston University School of MedicineBostonMAUSA
| | - Daehee Hwang
- Department of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Hoon Ryu
- Veteran's Affairs Boston Healthcare SystemBostonMAUSA,Department of NeurologyBoston University Alzheimer’s Disease CenterBoston University School of MedicineBostonMAUSA,Center for NeuromedicineBrain Science InstituteKorea Institute of Science and TechnologySeoulSouth Korea
| |
Collapse
|
55
|
Nahálková J. Linking TPPII to the protein interaction and signalling networks. Comput Biol Chem 2020; 87:107291. [PMID: 32702546 DOI: 10.1016/j.compbiolchem.2020.107291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/21/2020] [Accepted: 05/22/2020] [Indexed: 01/18/2023]
Abstract
Tripeptidyl peptidase II (TPPII) is primarily considered a house-keeping exopeptidase, which contributes to the functions of the ubiquitin-proteasome system by the maintenance of the cellular amino acid homeostasis. Although functionally well-characterised in vitro and using the mammalian cell models, less is known about the molecular mechanisms of its involvement in the signalling and metabolic pathways, which mediate its cellular functions. The present protein-protein interaction network analysis identified these mechanisms involved in the adaptive and innate immunity, the metabolism of the glucose, cancer cell growth, apoptosis, cell cycle and DNA damage responses. The interaction network constructed based on the publicly available protein-protein interaction data was extended by the application GeneMania, which was further used for the pathway enrichment, the protein function prediction and the protein node prioritisation analysis. The analysis suggested that the molecular mechanisms linked to the adaptive and innate immunity (ID, Kit receptor, BCR, IL-2 and G-CSF signalling; the regulation of NFκB), the aerobic glycolysis (ID and IL-2 signalling), tumorigenesis (TGF-β and p53 signalling; the top priority nodes MAPKs, mTOR regulation), diabetes (Kit receptor signalling; the top priority node GSK3β) and neurodegeneration (the control of mTOR and Aβ peptide degradation) are controlling the resulting TPPII interaction network. The uncharacterized interactions with two lung cancer suppressors (DOK3, DENND2D), a protein involved in the increased risk of the lung cancer in smokers (CYP1A1) and a protein implicated in asthmatic reactions (CHIA) suggest potential roles of TPPII in the lung cancer pathology. The interactions with methyltransferase CARNMT1, which modifies di- and tripeptides and the xenobiotic processing enzyme CYP1A1, are additional candidates for the breakthrough in new functions discovery of TPPII.
Collapse
Affiliation(s)
- Jarmila Nahálková
- Biochemworld Co., Biochemistry, Molecular & Cell Biology Unit, Snickar-Anders väg 17, 74394, Skyttorp, Uppsala County, Sweden.
| |
Collapse
|
56
|
Zorzan I, Pellegrini M, Arboit M, Incarnato D, Maldotti M, Forcato M, Tagliazucchi GM, Carbognin E, Montagner M, Oliviero S, Martello G. The transcriptional regulator ZNF398 mediates pluripotency and epithelial character downstream of TGF-beta in human PSCs. Nat Commun 2020; 11:2364. [PMID: 32398665 PMCID: PMC7217929 DOI: 10.1038/s41467-020-16205-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) have the capacity to give rise to all differentiated cells of the adult. TGF-beta is used routinely for expansion of conventional hPSCs as flat epithelial colonies expressing the transcription factors POU5F1/OCT4, NANOG, SOX2. Here we report a global analysis of the transcriptional programme controlled by TGF-beta followed by an unbiased gain-of-function screening in multiple hPSC lines to identify factors mediating TGF-beta activity. We identify a quartet of transcriptional regulators promoting hPSC self-renewal including ZNF398, a human-specific mediator of pluripotency and epithelial character in hPSCs. Mechanistically, ZNF398 binds active promoters and enhancers together with SMAD3 and the histone acetyltransferase EP300, enabling transcription of TGF-beta targets. In the context of somatic cell reprogramming, inhibition of ZNF398 abolishes activation of pluripotency and epithelial genes and colony formation. Our findings have clear implications for the generation of bona fide hPSCs for regenerative medicine.
Collapse
Affiliation(s)
- Irene Zorzan
- Department of Molecular Medicine, Medical School, University of Padua, 35121, Padua, Italy
| | - Marco Pellegrini
- Department of Molecular Medicine, Medical School, University of Padua, 35121, Padua, Italy.,UCL Great Ormond Street Institute of Child Health, Developmental Biology and Cancer, Stem Cells and Regenerative Medicine, 30 Guilford Street, WC1N 1EH, London, UK
| | - Mattia Arboit
- Department of Molecular Medicine, Medical School, University of Padua, 35121, Padua, Italy
| | - Danny Incarnato
- Department of Life Sciences and Systems Biology and Molecular Biotechnology Center (MCB), University of Turin, 10126, Turin, Italy.,Italian Institute for Genomic Medicine (IIGM), 10060, Candiolo (TO), Italy.,Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Mara Maldotti
- Department of Life Sciences and Systems Biology and Molecular Biotechnology Center (MCB), University of Turin, 10126, Turin, Italy.,Italian Institute for Genomic Medicine (IIGM), 10060, Candiolo (TO), Italy
| | - Mattia Forcato
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Guidantonio Malagoli Tagliazucchi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy.,UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Darwin Building, WC1E 6BT, London, UK
| | - Elena Carbognin
- Department of Molecular Medicine, Medical School, University of Padua, 35121, Padua, Italy
| | - Marco Montagner
- Department of Molecular Medicine, Medical School, University of Padua, 35121, Padua, Italy
| | - Salvatore Oliviero
- Department of Life Sciences and Systems Biology and Molecular Biotechnology Center (MCB), University of Turin, 10126, Turin, Italy. .,Italian Institute for Genomic Medicine (IIGM), 10060, Candiolo (TO), Italy.
| | - Graziano Martello
- Department of Molecular Medicine, Medical School, University of Padua, 35121, Padua, Italy.
| |
Collapse
|
57
|
Gadomski S, Singh SK, Singh S, Sarkar T, Klarmann KD, Berenschot M, Seaman S, Jakubison B, Gudmundsson KO, Lockett S, Keller JR. Id1 and Id3 Maintain Steady-State Hematopoiesis by Promoting Sinusoidal Endothelial Cell Survival and Regeneration. Cell Rep 2020; 31:107572. [PMID: 32348770 PMCID: PMC8459380 DOI: 10.1016/j.celrep.2020.107572] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/19/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
Investigating mechanisms that regulate endothelial cell (EC) growth and survival is important for understanding EC homeostasis and how ECs maintain stem cell niches. We report here that targeted loss of Id genes in adult ECs results in dilated, leaky sinusoids and a pro-inflammatory state that increases in severity over time. Disruption in sinusoidal integrity leads to increased hematopoietic stem cell (HSC) proliferation, differentiation, migration, and exhaustion. Mechanistically, sinusoidal ECs (SECs) show increased apoptosis because of reduced Bcl2-family gene expression following Id gene ablation. Furthermore, Id1-/-Id3-/- SECs and upstream type H vessels show increased expression of cyclin-dependent kinase inhibitors p21 and p27 and impaired ability to proliferate, which is rescued by reducing E2-2 expression. Id1-/-Id3-/- mice do not survive sublethal irradiation because of impaired vessel regeneration and hematopoietic failure. Thus, Id genes are required for the survival and regeneration of BM SECs during homeostasis and stress to maintain HSC development.
Collapse
Affiliation(s)
- Stephen Gadomski
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Satyendra K Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Shweta Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Tanmoy Sarkar
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Kimberly D Klarmann
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Maximillian Berenschot
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Steven Seaman
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Brad Jakubison
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Kristbjorn O Gudmundsson
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jonathan R Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
| |
Collapse
|
58
|
Mondal B, Jin H, Kallappagoudar S, Sedkov Y, Martinez T, Sentmanat MF, Poet GJ, Li C, Fan Y, Pruett-Miller SM, Herz HM. The histone deacetylase complex MiDAC regulates a neurodevelopmental gene expression program to control neurite outgrowth. eLife 2020; 9:57519. [PMID: 32297854 PMCID: PMC7192582 DOI: 10.7554/elife.57519] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
The mitotic deacetylase complex (MiDAC) is a recently identified histone deacetylase (HDAC) complex. While other HDAC complexes have been implicated in neurogenesis, the physiological role of MiDAC remains unknown. Here, we show that MiDAC constitutes an important regulator of neural differentiation. We demonstrate that MiDAC functions as a modulator of a neurodevelopmental gene expression program and binds to important regulators of neurite outgrowth. MiDAC upregulates gene expression of pro-neural genes such as those encoding the secreted ligands SLIT3 and NETRIN1 (NTN1) by a mechanism suggestive of H4K20ac removal on promoters and enhancers. Conversely, MiDAC inhibits gene expression by reducing H3K27ac on promoter-proximal and -distal elements of negative regulators of neurogenesis. Furthermore, loss of MiDAC results in neurite outgrowth defects that can be rescued by supplementation with SLIT3 and/or NTN1. These findings indicate a crucial role for MiDAC in regulating the ligands of the SLIT3 and NTN1 signaling axes to ensure the proper integrity of neurite development.
Collapse
Affiliation(s)
- Baisakhi Mondal
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Hongjian Jin
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Satish Kallappagoudar
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Yurii Sedkov
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Tanner Martinez
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Monica F Sentmanat
- Genome Engineering & iPS Center, Department of Genetics, Washington University, St. Louis, United States
| | - Greg J Poet
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Shondra M Pruett-Miller
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Hans-Martin Herz
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| |
Collapse
|
59
|
Sakamoto S, Tateya T, Omori K, Kageyama R. Idgenes are required for morphogenesis and cellular patterning in the developing mammalian cochlea. Dev Biol 2020; 460:164-175. [PMID: 31843520 DOI: 10.1016/j.ydbio.2019.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 10/25/2022]
Abstract
Inhibitor of differentiation and DNA-binding (Id) proteins, Id1 to Id4, function in the regulation of cellular proliferation and differentiation. Id proteins have been shown to interact with bHLH proteins and other proteins involved in regulating cellular proliferation and differentiation, suggesting a widespread regulatory function. Id1-3 are known to be expressed in the prosensory domain of developing cochlea. However, the roles of Id genes in cochlear development are not fully elucidated. The deficiency of any of the Id1-3 genes individually has little effect on the cochlear development, and therefore the functional redundancy among these genes have been presumed to explain the absence of phenotype. Here, we show that conditional knockout of Id1/2/3 genes (Id TKO) causes major defects in morphogenesis and cellular patterning in the development of mammalian cochlea. Id TKO cochlea was 82% shorter than control, and both decreased proliferation and increased cell death caused the hypomorph. Sox2-positive prosensory domain was formed in Id TKO cochlea, but the formation of the medial-lateral (central-peripheral) axis was disturbed; the boundary between the medial and lateral compartments in the prosensory domain was partially doubled; the number of inner hair cells per unit length increased, and the number of outer hair cells decreased. Furthermore, the lateral non-sensory compartment expressing Bmp4 and Lmo3 was missing. Thus, the patterning of the lateral epithelium was more affected than the medial epithelium. These results suggested that Id genes are crucial for morphogenesis of the cochlea duct and patterning of the lateral epithelium in the developing cochlea. Further analyses by quantitative RT-PCR and immunostaining using cochlear explants with a Bmp pathway inhibitor revealed that the Bmp-Id pathway originates from the lateral non-sensory compartment and promotes outer hair cell differentiation.
Collapse
Affiliation(s)
- Susumu Sakamoto
- Department of Otolaryngology, Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan; Kyoto University of Advanced Science, Kyoto, 615-8577, Japan
| | - Tomoko Tateya
- Department of Otolaryngology, Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan; Kyoto University of Advanced Science, Kyoto, 615-8577, Japan; Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan; Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan; Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
60
|
Saravanan S, Vimalraj S, Pavani K, Nikarika R, Sumantran VN. Intussusceptive angiogenesis as a key therapeutic target for cancer therapy. Life Sci 2020; 252:117670. [PMID: 32298741 DOI: 10.1016/j.lfs.2020.117670] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/20/2022]
Abstract
Deregulation of angiogenesis is a key reason for tumor growth and progression. Several anti-angiogenic drugs in clinical practice attempt to normalize abnormal tumor vasculature. Unfortunately, these drugs are ineffective due to the development of resistance in patients after drug holidays. A sizable literature suggests that resistance to these anti-angiogenic drugs occurs due to various compensatory mechanisms of tumor angiogenesis. Therefore, we describe different compensatory mechanisms of tumor angiogenesis, and explain why intussusceptive angiogenesis (IA), is a crucial mechanism of compensatory angiogenesis in tumors which resist anti-VEGF (vascular endothelial growth factor) therapies. IA is often overlooked due to the scarcity of experimental models. Therefore, we examine data from existing experimental models and our novel ex-ovo model of angiogenesis in chick embryos, and explain the important genes and signaling pathways driving IA. Using bio-informatic analyses of major genes regulating conventional sprouting angiogenesis (SA) and intussusceptive angiogenesis, we provide fresh insights on the 'angiogenic switch' which regulates the transition from SA to IA. Finally, we examine the interplay between molecules regulating SA, IA, and molecules known to promote tumor progression. Based on these analyses, we conclude that intussusceptive angiogenesis (IA) is a promising therapeutic target for developing effective anti-cancer treatment regimes.
Collapse
Affiliation(s)
- Sekaran Saravanan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), Department of Bioengineering, School of Chemical and Biotechnology, SASTRA University, Thanjavur 613 401, Tamil Nadu, India
| | - Selvaraj Vimalraj
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India.
| | - Koka Pavani
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India
| | - Ramesh Nikarika
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India
| | - Venil N Sumantran
- Abdul Kalam Center for Innovation and Entrepreneurship, Dr. MGR Educational & Research Institute, Maduravoyal, Chennai 600095, India
| |
Collapse
|
61
|
Panahipour L, Biasi MD, Bokor TS, Thajer A, Haiden N, Gruber R. Milk lactoperoxidase decreases ID1 and ID3 expression in human oral squamous cell carcinoma cell lines. Sci Rep 2020; 10:5836. [PMID: 32246075 PMCID: PMC7125221 DOI: 10.1038/s41598-020-62390-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 03/12/2020] [Indexed: 01/07/2023] Open
Abstract
Milk consumption may modify the risk of squamous cell carcinoma. The role of milk to modulate the gene expression in oral squamous cell carcinoma cells has not been investigated so far. Here, HSC2 oral squamous carcinoma cells were exposed to an aqueous fraction of human milk and a whole-genome array was performed. Among the genes that were significantly reduced by human and cow milk were the DNA-binding protein inhibitor 1 (ID1), ID3 and Distal-Less Homeobox 2 (DLX2) in HSC2 cells. Also, in TR146 oral squamous carcinoma cells, there was a tendency towards a decreased gene expression. Upon size fractionation, lactoperoxidase but not lactoferrin and osteopontin was identified to reduce ID1 and ID3 in HSC2 cells. Dairy products and hypoallergenic infant formula failed to decrease the respective genes. These data suggest that milk can reduce the expression of transcription factors in oral squamous carcinoma cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Maria De Biasi
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Theresa Sophia Bokor
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Alexandra Thajer
- Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Nadja Haiden
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria. .,Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010, Bern, Switzerland. .,Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200, Vienna, Austria.
| |
Collapse
|
62
|
Zhao Z, Bo Z, Gong W, Guo Y. Inhibitor of Differentiation 1 (Id1) in Cancer and Cancer Therapy. Int J Med Sci 2020; 17:995-1005. [PMID: 32410828 PMCID: PMC7211148 DOI: 10.7150/ijms.42805] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The inhibitor of DNA binding (Id) proteins are regulators of cell cycle and cell differentiation. Of all Id family proteins, Id1 is mostly linked to tumorigenesis, cellular senescence as well as cell proliferation and survival. Id1 is a stem cell-like gene more than a classical oncogene. Id1 is overexpressed in numerous types of cancers and exerts its promotion effect to these tumors through different pathways. Briefly, Id1 was found significantly correlated with EMT-related proteins, K-Ras signaling, EGFR signaling, BMP signaling, PI3K/Akt signaling, WNT and SHH signaling, c-Myc signaling, STAT3 signaling, RK1/2 MAPK/Egr1 pathway and TGF-β pathway, etc. Id1 has potent effect on facilitating tumorous angiogenesis and metastasis. Moreover, high expression of Id1 plays a facilitating role in the development of drug resistance, including chemoresistance, radiation resistance and resistance to drugs targeting angiogenesis. However, controversial results were also obtained. Overall, Id1 represent a promising target of anti-tumor therapeutics based on its potent promotion effect to cancer. Numerous drugs were found exerting their anti-tumor function through Id1-related signaling pathways, such as fucoidan, berberine, tetramethylpyrazine, crizotinib, cannabidiol and vinblastine.
Collapse
Affiliation(s)
- Zhengxiao Zhao
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Zhiyuan Bo
- The Second Department of Biliary Tract Surgery, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| | - Weiyi Gong
- The Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, PR China
| | - Yong Guo
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
63
|
Wojnarowicz PM, Lima E Silva R, Ohnaka M, Lee SB, Chin Y, Kulukian A, Chang SH, Desai B, Garcia Escolano M, Shah R, Garcia-Cao M, Xu S, Kadam R, Goldgur Y, Miller MA, Ouerfelli O, Yang G, Arakawa T, Albanese SK, Garland WA, Stoller G, Chaudhary J, Norton L, Soni RK, Philip J, Hendrickson RC, Iavarone A, Dannenberg AJ, Chodera JD, Pavletich N, Lasorella A, Campochiaro PA, Benezra R. A Small-Molecule Pan-Id Antagonist Inhibits Pathologic Ocular Neovascularization. Cell Rep 2019; 29:62-75.e7. [PMID: 31577956 PMCID: PMC6896334 DOI: 10.1016/j.celrep.2019.08.073] [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: 08/14/2018] [Revised: 08/09/2019] [Accepted: 08/23/2019] [Indexed: 02/01/2023] Open
Abstract
Id helix-loop-helix (HLH) proteins (Id1-4) bind E protein bHLH transcription factors, preventing them from forming active transcription complexes that drive changes in cell states. Id proteins are primarily expressed during development to inhibit differentiation, but they become re-expressed in adult tissues in diseases of the vasculature and cancer. We show that the genetic loss of Id1/Id3 reduces ocular neovascularization in mouse models of wet age-related macular degeneration (AMD) and retinopathy of prematurity (ROP). An in silico screen identifies AGX51, a small-molecule Id antagonist. AGX51 inhibits the Id1-E47 interaction, leading to ubiquitin-mediated degradation of Ids, cell growth arrest, and reduced viability. AGX51 is well-tolerated in mice and phenocopies the genetic loss of Id expression in AMD and ROP models by inhibiting retinal neovascularization. Thus, AGX51 is a first-in-class compound that antagonizes an interaction formerly considered undruggable and that may have utility in the management of multiple diseases.
Collapse
Affiliation(s)
- Paulina M Wojnarowicz
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raquel Lima E Silva
- Departments of Ophthalmology and Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Masayuki Ohnaka
- Departments of Ophthalmology and Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sang Bae Lee
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Yvette Chin
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anita Kulukian
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sung-Hee Chang
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bina Desai
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marta Garcia Escolano
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Riddhi Shah
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marta Garcia-Cao
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sijia Xu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rashmi Kadam
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yehuda Goldgur
- Structural Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Meredith A Miller
- Structural Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ouathek Ouerfelli
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Guangli Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tsutomu Arakawa
- Alliance Protein Laboratories, a Division of KBI Biopharma, San Diego, CA 92121, USA
| | - Steven K Albanese
- Computational Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Glenn Stoller
- Ophthalmic Consultants of Long Island, Lynbrook, NY 11563, USA
| | - Jaideep Chaudhary
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Larry Norton
- Evelyn H. Lauder Breast Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rajesh Kumar Soni
- Proteomics & Microchemistry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - John Philip
- Proteomics & Microchemistry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ronald C Hendrickson
- Proteomics & Microchemistry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Antonio Iavarone
- Department of Neurology, Department of Pathology, Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - John D Chodera
- Computational Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nikola Pavletich
- Structural Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Lasorella
- Department of Pediatrics, Department of Pathology, Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter A Campochiaro
- Departments of Ophthalmology and Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
64
|
Krentz NAJ, Lee MYY, Xu EE, Sproul SLJ, Maslova A, Sasaki S, Lynn FC. Single-Cell Transcriptome Profiling of Mouse and hESC-Derived Pancreatic Progenitors. Stem Cell Reports 2019; 11:1551-1564. [PMID: 30540962 PMCID: PMC6294286 DOI: 10.1016/j.stemcr.2018.11.008] [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: 04/18/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 01/06/2023] Open
Abstract
Human embryonic stem cells (hESCs) are a potential unlimited source of insulin-producing β cells for diabetes treatment. A greater understanding of how β cells form during embryonic development will improve current hESC differentiation protocols. All pancreatic endocrine cells, including β cells, are derived from Neurog3-expressing endocrine progenitors. This study characterizes the single-cell transcriptomes of 6,905 mouse embryonic day (E) 15.5 and 6,626 E18.5 pancreatic cells isolated from Neurog3-Cre; Rosa26mT/mG embryos, allowing for enrichment of endocrine progenitors (yellow; tdTomato + EGFP) and endocrine cells (green; EGFP). Using a NEUROG3-2A-eGFP CyT49 hESC reporter line (N5-5), 4,462 hESC-derived GFP+ cells were sequenced. Differential expression analysis revealed enrichment of markers that are consistent with progenitor, endocrine, or previously undescribed cell-state populations. This study characterizes the single-cell transcriptomes of mouse and hESC-derived endocrine progenitors and serves as a resource (https://lynnlab.shinyapps.io/embryonic_pancreas) for improving the formation of functional β-like cells from hESCs. Single-cell transcriptome of embryonic mouse pancreas and hESC-derived cells Identification of novel cell types during mouse pancreas development Pseudotime analysis reveals developmental trajectories of endocrine cell lineage hESC-derived endocrine cells resemble immature β cells
Collapse
Affiliation(s)
- Nicole A J Krentz
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28(th) Avenue West, Vancouver, BC V5Z4H4, Canada.
| | - Michelle Y Y Lee
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Eric E Xu
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28(th) Avenue West, Vancouver, BC V5Z4H4, Canada
| | - Shannon L J Sproul
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28(th) Avenue West, Vancouver, BC V5Z4H4, Canada
| | - Alexandra Maslova
- Graduate Program in Bioinformatics, University of British Columbia, 100-570 7(th) Avenue West, Vancouver, BC V5Z 4S6, Canada
| | - Shugo Sasaki
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28(th) Avenue West, Vancouver, BC V5Z4H4, Canada
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28(th) Avenue West, Vancouver, BC V5Z4H4, Canada.
| |
Collapse
|
65
|
Li H, Miki T, Almeida GMD, Hanashima C, Matsuzaki T, Kuo CJ, Watanabe N, Noda M. RECK in Neural Precursor Cells Plays a Critical Role in Mouse Forebrain Angiogenesis. iScience 2019; 19:559-571. [PMID: 31445376 PMCID: PMC6713797 DOI: 10.1016/j.isci.2019.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/28/2019] [Accepted: 08/05/2019] [Indexed: 01/10/2023] Open
Abstract
RECK in neural precursor cells (NPCs) was previously found to support Notch-dependent neurogenesis in mice. On the other hand, recent studies implicate RECK in endothelial cells (ECs) in WNT7-triggered canonical WNT signaling essential for brain angiogenesis. Here we report that RECK in NPCs is also critical for brain angiogenesis. When Reck is inactivated in Foxg1-positive NPCs, mice die shortly after birth with hemorrhage in the forebrain, with angiogenic sprouts stalling at the periphery and forming abnormal aggregates reminiscent of those in EC-selective Reck knockout mice and Wnt7a/b-deficient mice. The hemorrhage can be pharmacologically suppressed by lithium chloride. An effect of RECK in WNT7-producing cells to enhance canonical WNT-signaling in reporter cells is detectable in mixed culture but not with conditioned medium. Our findings suggest that NPC-expressed RECK has a non-cell-autonomous function to promote forebrain angiogenesis through contact-dependent enhancement of WNT signaling in ECs, implying possible involvement of RECK in neurovascular coupling. Mice lacking RECK in Foxg1-positive neural precursor cells die shortly after birth These mice show vascular defects similar to those in mice lacking endothelial RECK The vascular phenotype can be suppressed by LiCl, an activator of WNT signaling RECK in WNT7-producing cell enhances contact-dependent WNT signaling in adjacent cells
Collapse
Affiliation(s)
- Huiping Li
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takao Miki
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Glícia Maria de Almeida
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Carina Hanashima
- Department of Biology, Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Tomoko Matsuzaki
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Calvin J Kuo
- Stanford University School of Medicine, Department of Medicine, Division of Hematology, Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA 94305, USA
| | - Naoki Watanabe
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Pharmacology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Makoto Noda
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
66
|
Avecilla V. Effect of Transcriptional Regulator ID3 on Pulmonary Arterial Hypertension and Hereditary Hemorrhagic Telangiectasia. Int J Vasc Med 2019; 2019:2123906. [PMID: 31380118 PMCID: PMC6657613 DOI: 10.1155/2019/2123906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/26/2019] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) can be discovered in patients who have a loss of function mutation of activin A receptor-like type 1 (ACVRL1) gene, a bone morphogenetic protein (BMP) type 1 receptor. Additionally, ACVRL1 mutations can lead to hereditary hemorrhagic telangiectasia (HHT), also known as Rendu-Osler-Weber disease, an autosomal dominant inherited disease that results in mucocutaneous telangiectasia and arteriovenous malformations (AVMs). Transcriptional regulator Inhibitor of DNA-Binding/Differentiation-3 (ID3) has been demonstrated to be involved in both PAH and HTT; however, the role of its overlapping molecular mechanistic effects has yet to be seen. This review will focus on the existing understanding of how ID3 may contribute to molecular involvement and perturbations thus altering both PAH and HHT outcomes. Improved understanding of how ID3 mediates these pathways will likely provide knowledge in the inhibition and regulation of these diseases through targeted therapies.
Collapse
Affiliation(s)
- Vincent Avecilla
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA
- Celgene Corporation, Summit, NJ 07901, USA
| |
Collapse
|
67
|
Malaguti M, Migueles RP, Blin G, Lin CY, Lowell S. Id1 Stabilizes Epiblast Identity by Sensing Delays in Nodal Activation and Adjusting the Timing of Differentiation. Dev Cell 2019; 50:462-477.e5. [PMID: 31204172 PMCID: PMC6706657 DOI: 10.1016/j.devcel.2019.05.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/04/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
Controlling responsiveness to prevailing signals is critical for robust transitions between cell states during development. For example, fibroblast growth factor (FGF) drives naive pluripotent cells into extraembryonic lineages before implantation but sustains pluripotency in primed cells of the post-implantation epiblast. Nanog supports pluripotency in naive cells, while Nodal supports pluripotency in primed cells, but the handover from Nanog to Nodal does not proceed seamlessly, opening up the risk of aberrant differentiation if FGF is activated before Nodal. Here, we report that Id1 acts as a sensor to detect delays in Nodal activation after the downregulation of Nanog. Id1 then suppresses FGF activity to delay differentiation. Accordingly, Id1 is not required for naive or primed pluripotency but rather stabilizes epiblast identity during the transition between these states. These findings help explain how development proceeds robustly in the face of imprecise signals and highlight the importance of mechanisms that stabilize cell identity during developmental transitions.
Collapse
Affiliation(s)
- Mattias Malaguti
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, the University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Rosa Portero Migueles
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, the University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Guillaume Blin
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, the University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Chia-Yi Lin
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, the University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, the University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
68
|
Zhao Z, Liu B, Sun J, Lu L, Liu L, Qiu J, Li Q, Yan C, Jiang S, Mohammadtursun N, Ma W, Li M, Dong J, Gong W. Scutellaria Flavonoids Effectively Inhibit the Malignant Phenotypes of Non-small Cell Lung Cancer in an Id1-dependent Manner. Int J Biol Sci 2019; 15:1500-1513. [PMID: 31337979 PMCID: PMC6643150 DOI: 10.7150/ijbs.33146] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/23/2019] [Indexed: 12/17/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer death in the world. Inhibitor of differentiation 1 (Id1) is overexpressed in NSCLC and involved in promoting its progression and metastasis. Identifying natural compounds targeting Id1 may have utility in NSCLC treatment. Here, we sought to determine whether the anti-tumor activities of Scutellaria flavonoids (SFs) were related to Id1. We reported that three SFs (baicalin, baicalein and wogonin) exhibited strong antitumor activity in NSCLC cells in vitro and in vivo. Id1 played a pivotal role on blockage of migration and invasion by SFs. Abrogation of invasion and migration mediated by baicalin, baicalein and wogonin were totally abolished by ectopic overexpression of Id1. Mechanistically, baicalin, baicalein and wogonin activated Rap1-GTP binding and dephosphorylated Akt and Src by suppressing a7nAChR, consequently triggering inhibition of Id1. Then attenuation of its downstream mediators, VEGF-A, N-cadherin, vimentin, combined with augment of E-cadherin led to the blockage of proliferation, EMT and angiogenesis of NSCLC. Overall, our data shed light on heretofore-undescribed role of SFs as modulators of Id1, which may be a useful strategy in the treatment of NSCLC.
Collapse
Affiliation(s)
- Zhengxiao Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Lumei Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Jian Qiu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Qiuping Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Chen Yan
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Shan Jiang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Nabijan Mohammadtursun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Wenjuan Ma
- Department of dermatology, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Mihui Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China.,Institutes of Integrative Medicine, Fudan University, Shanghai, PR China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Weiyi Gong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| |
Collapse
|
69
|
Richter A, Alexdottir MS, Magnus SH, Richter TR, Morikawa M, Zwijsen A, Valdimarsdottir G. EGFL7 Mediates BMP9-Induced Sprouting Angiogenesis of Endothelial Cells Derived from Human Embryonic Stem Cells. Stem Cell Reports 2019; 12:1250-1259. [PMID: 31155507 PMCID: PMC6565989 DOI: 10.1016/j.stemcr.2019.04.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 12/21/2022] Open
Abstract
Human embryonic stem cells (hESCs) are instrumental in characterizing the molecular mechanisms of human vascular development and disease. Bone morphogenetic proteins (BMPs) play a pivotal role in cardiovascular development in mice, but their importance for vascular cells derived from hESCs has not yet been fully explored. Here, we demonstrate that BMP9 promotes, via its receptor ALK1 and SMAD1/5 activation, sprouting angiogenesis of hESC-derived endothelial cells. We show that the secreted angiogenic factor epidermal growth factor-like domain 7 (EGFL7) is a downstream target of BMP9-SMAD1/5-mediated signaling, and that EGFL7 promotes expansion of endothelium via interference with NOTCH signaling, activation of ERK, and remodeling of the extracellular matrix. CRISPR/Cas9-mediated deletion of EGFL7 highlights the critical role of EGFL7 in BMP9-induced endothelial sprouting and the promotion of angiogenesis. Our study illustrates the complex role of the BMP family in orchestrating hESC vascular development and endothelial sprouting. BMP9/ALK1 signaling induces sprouting of hESC-derived endothelial cells EGFL7 mediates BMP9-induced sprouting angiogenesis of hESC-derived endothelial cells EGFL7 inhibits the NOTCH pathway and activates the ERK pathway in HUVECs EGFL7 affects the extracellular matrix in HUVECs
Collapse
Affiliation(s)
- Anne Richter
- Department of Biochemistry and Molecular Biology, BioMedical Center, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| | - Marta S Alexdottir
- Department of Anatomy, BioMedical Center, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland
| | - Svala H Magnus
- Department of Biochemistry and Molecular Biology, BioMedical Center, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| | - Tobias R Richter
- Department of Anatomy, BioMedical Center, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland
| | - Masato Morikawa
- Ludwig Institute for Cancer Research, Uppsala University, 751 24 Uppsala, Sweden
| | - An Zwijsen
- VIB-KU Leuven Center for Brain and Disease Research, ON4 Herestraat 49, Box 602, 3000 Leuven, Belgium; KU Leuven Department of Cardiovascular Sciences, ON4 Herestraat 49, Box 911, 3000 Leuven, Belgium
| | - Gudrun Valdimarsdottir
- Department of Biochemistry and Molecular Biology, BioMedical Center, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland; Department of Anatomy, BioMedical Center, University of Iceland, Sturlugata 8, 101 Reykjavik, Iceland.
| |
Collapse
|
70
|
Hu L, Pu Q, Zhang Y, Ma Q, Li G, Li X. Expansion and maintenance of primary corneal epithelial stem/progenitor cells by inhibition of TGFβ receptor I-mediated signaling. Exp Eye Res 2019; 182:44-56. [PMID: 30914160 DOI: 10.1016/j.exer.2019.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 01/10/2023]
Abstract
Transforming growth factor β (TGFβ) signaling is one of the most important signaling pathways regulating cell behavior in ocular tissues. Its functions are mainly linked to tissue fibrosis and inflammatory responses in ophthalmology. In epithelial cells, however, the growth inhibitory activity of TGFβ was reported in both non-ocular and ocular tissues. Since TGFβ is a bifunctional regulator that either inhibits or stimulates cell proliferation according to the specific context, we examined the effect of inhibition of TGFβ receptor (TβR) I-mediated signaling on primary corneal epithelial cells (CECs) in serum- and feeder-free conditions. The mouse CECs were isolated from the eyeballs of 6-8 weeks old female C57BL/6 mice using dispase and trypsin separately, cultivated in defined Keratinocyte serum-free medium (KSFM) with supplements (the complete medium) without feeder layer. Cells were divided into three groups, those cultured in complete medium additionally supplemented with 10 μM SB-431542, a specific inhibitor of TβR-I, were SB-CECs; those cultured in complete medium additionally supplemented with 10 ng/ml SRI-011381, a TGF-beta signaling agonist, were SRI-CECs; those cultured in complete medium without SB-431542 or SRI-011381 were control CECs. The growth rate and morphology were analyzed by light microscopy. The identity and stemness of cells was investigated through marker staining of p63, inhibitor of differentiation 1 (ID1), cytokeratin 12 (K12), cytokeratin 14 (K14), PAX6, pSmad3, alpha smooth muscle Actin (αSMA) and E-cadherin (E-cad); Real-time quantitative (RT-PCR) analysis of p63; Western blot analysis of ID1; as well as colony forming assay, sphere forming assay, healing wound in vitro assay and air-lifting interface assay. The results showed SB-CECs subcultured steadily, achieved sustained expansion, and expanded almost thrice faster than control CECs. Expanded SB-CECs exhibited smaller and more compact morphology, up-regulated p63 and ID1, as well as better performed colony-forming capacity, sphere-forming capacity, in vitro wound healing capacity, and the capacity to stratify and differentiate on air-lifting interface. Preliminary tests on human limbal epithelial cells (HLECs) showed the same results as mouse CECs. Interestingly, the ID1 expression pattern was almost identical to p63, the typical marker for corneal epithelial stem/progenitor cell (CESC/CEPC), in cultured CECs and normal corneal sections. Since ID1 has been proven to be regulated negatively by TGFβ signaling in epithelial cells and plays a role in blocking cell differentiation, its derepression by TβR-I inhibitor could be, at least in part, the underlying cause of CESC/CEPC expansion and the synchronously up-regulated expression of p63 in SB-CECs. In conclusion, inhibition of TβR-I-mediated signaling, CESCs/CEPCs achieved efficient long-term expansion in a feeder- and serum-free condition in vitro. And derepression of ID1 could be the underlying cause. Meanwhile, ID1 could serve as a marker for CESC/CEPC. These results may advance the basic and clinical CESC/CEPC research.
Collapse
Affiliation(s)
- Lihua Hu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qi Pu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yaoli Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qian Ma
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xinyu Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
71
|
Shams R, Geranpayeh L, Omrani MD, Ghafouri-Fard S. Expression analysis of Inhibitor Of DNA Binding 1 (ID-1) gene in breast cancer. Hum Antibodies 2019; 27:129-134. [PMID: 30856107 DOI: 10.3233/hab-180358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND ID-1 gene codes for a helix-loop-helix (HLH) protein that inhibits the DNA binding and transcriptional activation function of these proteins. METHODS We analyzed ID-1 expression in microarray and RNA Sequencing databases as well as 61 breast cancer tissues compared with adjacent non-cancerous tissues (ANCTs). RESULTS Expression analysis of ID-1 gene in two microarray datasets and RNA sequencing data showed down-regulation of ID-1 in tumoral tissues compared with normal tissues. However, ID-1 expression analysis in tumoral tissues and ANCTs obtained from 61 patients revealed its over-expression in tumoral tissues. A negative association was detected between ID-1 expression levels and ER status. CONCLUSION ID-1 expression may be implicated in the pathogenesis of breast cancer especially in patient with ER negative status.
Collapse
Affiliation(s)
- Roshanak Shams
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lobat Geranpayeh
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
72
|
Manzo G. Similarities Between Embryo Development and Cancer Process Suggest New Strategies for Research and Therapy of Tumors: A New Point of View. Front Cell Dev Biol 2019; 7:20. [PMID: 30899759 PMCID: PMC6416183 DOI: 10.3389/fcell.2019.00020] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/05/2019] [Indexed: 12/25/2022] Open
Abstract
Here, I propose that cancer stem cells (CSCs) would be equivalent to para-embryonic stem cells (p-ESCs), derived from adult cells de-re-programmed to a ground state. p-ESCs would differ from ESCs by the absence of genomic homeostasis. A p-ESC would constitute the cancer cell of origin (i-CSC or CSC0), capable of generating an initial tumor, corresponding to a pre-implantation blastocyst. In a niche with proper signals, it would engraft as a primary tumor, corresponding to a post-implantation blastocyst. i-CSC progeny would form primary pluripotent and slow self-renewing CSCs (CSC1s), blocked in an undifferentiated state, corresponding to epiblast cells; CSC1s would be tumor-initiating cells (TICs). CSC1s would generate secondary CSCs (CSC2s), corresponding to hypoblast cells; CSC2s would be tumor growth cells (TGCs). CSC1s/CSC2s would generate tertiary CSCs (CSC3s), with a mesenchymal phenotype; CSC3s would be tumor migrating cells (TMCs), corresponding to mesodermal precursors at primitive streak. CSC3s with more favorable conditions (normoxia), by asymmetrical division, would differentiate into cancer progenitor cells (CPCs), and these into cancer differentiated cells (CDCs), thus generating a defined cell hierarchy and tumor progression, mimicking somito-histo-organogenesis. CSC3s with less favorable conditions (hypoxia) would delaminate and migrate as quiescent circulating micro-metastases, mimicking mesenchymal cells in gastrula morphogenetic movements. In metastatic niches, these CSC3s would install and remain dormant in the presence of epithelial/mesenchymal transition (EMT) signals and hypoxia. But, in the presence of mesenchymal/epithelial transition (MET) signals and normoxia, they would revert to self-renewing CSC1s, reproducing the same cell hierarchy of the primary tumor as macro-metastases. Further similarities between ontogenesis and oncogenesis involving crucial factors, such as ID, HSP70, HLA-G, CD44, LIF, and STAT3, are strongly evident at molecular, physiological and immunological levels. Much experimental data about these factors led to considering the cancer process as ectopic rudimentary ontogenesis, where CSCs have privileged immunological conditions. These would consent to CSC development in an adverse environment, just like an embryo, which is tolerated, accepted and favored by the maternal organism in spite of its paternal semi-allogeneicity. From all these considerations, novel research directions, potential innovative tumor therapy and prophylaxis strategies might, theoretically, result.
Collapse
Affiliation(s)
- Giovanni Manzo
- General Pathology, “La Sapienza” University of Rome, Retired, Botrugno, Italy
| |
Collapse
|
73
|
ACVR1 R206H cooperates with H3.1K27M in promoting diffuse intrinsic pontine glioma pathogenesis. Nat Commun 2019; 10:1023. [PMID: 30833574 PMCID: PMC6399349 DOI: 10.1038/s41467-019-08823-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/01/2019] [Indexed: 12/04/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an incurable pediatric brain tumor, with approximately 25% of DIPGs harboring activating ACVR1 mutations that commonly co-associate with H3.1K27M mutations. Here we show that in vitro expression of ACVR1 R206H with and without H3.1K27M upregulates mesenchymal markers and activates Stat3 signaling. In vivo expression of ACVR1 R206H or G328V with H3.1K27M and p53 deletion induces glioma-like lesions but is not sufficient for full gliomagenesis. However, in combination with PDGFA signaling, ACVR1 R206H and H3.1K27M significantly decrease survival and increase tumor incidence. Treatment of ACVR1 R206H mutant DIPGs with exogenous Noggin or the ACVR1 inhibitor LDN212854 significantly prolongs survival, with human ACVR1 mutant DIPG cell lines also being sensitive to LDN212854 treatment. Together, our results demonstrate that ACVR1 R206H and H3.1K27M promote tumor initiation, accelerate gliomagenesis, promote a mesenchymal profile partly due to Stat3 activation, and identify LDN212854 as a promising compound to treat DIPG.
Collapse
|
74
|
Sengupta D, Kar S. Deciphering the Dynamical Origin of Mixed Population during Neural Stem Cell Development. Biophys J 2019; 114:992-1004. [PMID: 29490258 DOI: 10.1016/j.bpj.2017.12.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/08/2017] [Accepted: 12/27/2017] [Indexed: 02/04/2023] Open
Abstract
Neural stem cells (NSCs) often give rise to a mixed population of cells during differentiation. However, the dynamical origin of these mixed states is poorly understood. In this article, our mathematical modeling study demonstrates that the bone morphogenetic protein 2 (BMP2) mediated disparate differentiation dynamics of NSCs in central and peripheral nervous systems essentially function through two distinct bistable switches that are mutually interconnected via a mushroom-like bifurcation. Stochastic simulations of the model reveal that the mixed population originates due to the existence of these bistable switching regulations and that the maintenance of such mixed states depends on the level of stochastic fluctuations of the system. It further demonstrates that due to extrinsic variability, cells in an NSC population can dynamically transit from mushroom to a unique isola kind of bifurcation state, which essentially extends the range of the BMP2-driven mixed population state during differentiation. Importantly, the model predicts that by individually altering the expression level of key regulatory proteins, the NSCs can be converted entirely to a preferred phenotype for BMP2 doses that previously resulted in a mixed population. Our findings show that efficient neuronal regeneration can be achieved by systematically maneuvering the differentiation dynamics.
Collapse
Affiliation(s)
- Dola Sengupta
- Department of Chemistry, IIT Bombay, Powai, Mumbai, India
| | - Sandip Kar
- Department of Chemistry, IIT Bombay, Powai, Mumbai, India.
| |
Collapse
|
75
|
Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and Regenerating the Lung Cell by Cell. Physiol Rev 2019; 99:513-554. [PMID: 30427276 DOI: 10.1152/physrev.00001.2018] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?
Collapse
Affiliation(s)
- Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| |
Collapse
|
76
|
Abstract
In this review from Murre, the evolution of HLH genes, the structures of HLH domains, and the elaborate activities of HLH proteins in multicellular life are discussed. Helix–loop–helix (HLH) proteins are dimeric transcription factors that control lineage- and developmental-specific gene programs. Genes encoding for HLH proteins arose in unicellular organisms >600 million years ago and then duplicated and diversified from ancestral genes across the metazoan and plant kingdoms to establish multicellularity. Hundreds of HLH proteins have been identified with diverse functions in a wide variety of cell types. HLH proteins orchestrate lineage specification, commitment, self-renewal, proliferation, differentiation, and homing. HLH proteins also regulate circadian clocks, protect against hypoxic stress, promote antigen receptor locus assembly, and program transdifferentiation. HLH proteins deposit or erase epigenetic marks, activate noncoding transcription, and sequester chromatin remodelers across the chromatin landscape to dictate enhancer–promoter communication and somatic recombination. Here the evolution of HLH genes, the structures of HLH domains, and the elaborate activities of HLH proteins in multicellular life are discussed.
Collapse
Affiliation(s)
- Cornelis Murre
- Division of Biological Sciences, University of California at San Diego, La Jolla, California 92903, USA
| |
Collapse
|
77
|
Avecilla V, Avecilla A. Inhibitor of DNA-Binding/Differentiation Proteins and Environmental Toxicants: Genomic Impact on the Onset of Depressive Dysfunction. ACTA ACUST UNITED AC 2019; 7:medsci7010007. [PMID: 30634536 PMCID: PMC6358799 DOI: 10.3390/medsci7010007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/18/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
The ongoing growth of the international occurrence of depression and its ability to co-occur with other serious medical disorders, such as heart disease, cancer, diabetes, and Parkinson’s disease, is a current public health problem. Inhibitor of DNA-Binding/Differentiation (ID) proteins are part of a group of transcriptional factors that have shown involvement in neurocognitive disorders and, therefore, may have influence on depressive disorders. Previously, it has been established that exposure to environmental estrogenic endocrine disruptors (EEDs), such as polychlorinated biphenyls (PCBs) and bisphenol A (BPA), have played an important role in the modulation of depressive disorders. Hence, based on many studies, we consider the impact of these environmental pollutants on the group of ID proteins and how they impact depressive outcomes. Improved knowledge of how ID proteins interact with depressive disorders, through EED exposure, will contribute essential evidence that can further benefit our public health community with innovative knowledge to prevent these types of mental illnesses.
Collapse
Affiliation(s)
- Vincent Avecilla
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA.
- Celgene Corporation, Summit, NJ 07901, USA.
| | - Andrea Avecilla
- Department of Clinical Psychology, University of Massachusetts Dartmouth, North Dartmouth, MA 02747, USA.
| |
Collapse
|
78
|
Gupta R, Cristea M, Frankel P, Ruel C, Chen C, Wang Y, Morgan R, Leong L, Chow W, Koczywas M, Koehler S, Lim D, Luu T, Martel C, McNamara M, Somlo G, Twardowski P, Yen Y, Idorenyi A, Raechelle T, Carroll M, Chung V. Randomized trial of oral cyclophosphamide versus oral cyclophosphamide with celecoxib for recurrent epithelial ovarian, fallopian tube, and primary peritoneal cancer. Cancer Treat Res Commun 2019; 21:100155. [PMID: 31279962 PMCID: PMC9018111 DOI: 10.1016/j.ctarc.2019.100155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 04/26/2023]
Abstract
BACKGROUND Oral metronomic chemotherapy, which has low toxicity, has demonstrated promising anti-tumor and anti-angiogenic properties that may lead to prolonged progression-free survival and improved response rates in patients with recurrent epithelial ovarian cancer (EOC). These effects may be enhanced by the co-administration of anti-angiogenic agents. METHODS We conducted a randomized phase II clinical trial to evaluate the therapeutic benefit of oral metronomic cyclophosphamide (CTX) alone and with the anti-angiogenic drug celecoxib in patients with gynecological malignancies. 52 patients were randomly assigned to two treatments arms: 50 mg oral CTX daily alone (Arm A) or with 400 mg celecoxib twice daily (Arm B). The primary endpoint was response rate. Secondary endpoints included toxicity, time to treatment failure, and overall survival. RESULTS In Arm A (n = 26), 3 patients (12%) had stable disease >6 months and 1 (4%) had a partial response. In Arm B, 5 (19%) had stable disease >6 months and 1 patient (4%) had a partial response. There were no significant between-group differences in overall survival (9.69 months [95% CI 3.84-13.18] vs. 12.55 months [6.67-17.61]) or in median time to treatment failure (1.84 months [1.68-2.76] vs. 1.92 months [1.64-5.22]). The most common adverse events were nausea, vomiting, and abdominal pain. CONCLUSIONS Oral metronomic CTX has activity with no major toxicities in heavily pretreated recurrent gynecological cancers and may be considered in patients with indolent disease. We did not observe any additional benefit of celecoxib treatment, though this may be due to small sample sizes.
Collapse
Affiliation(s)
- Rohan Gupta
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Mihaela Cristea
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Paul Frankel
- Information Sciences, City of Hope National Cancer Center, Duarte, CA, United States
| | - Christopher Ruel
- Information Sciences, City of Hope National Cancer Center, Duarte, CA, United States
| | - Chen Chen
- Clinical Informatics, City of Hope National Cancer Center, Duarte, CA, United States
| | - Yingyu Wang
- Clinical Informatics, City of Hope National Cancer Center, Duarte, CA, United States
| | - Robert Morgan
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Lucille Leong
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Warren Chow
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Marianna Koczywas
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Steve Koehler
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Dean Lim
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Thehang Luu
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Cynthia Martel
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Mark McNamara
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - George Somlo
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Przemyslaw Twardowski
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Yun Yen
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Amanam Idorenyi
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States
| | - Tinsley Raechelle
- Clinical Trial Office, City of Hope National Cancer Center, Duarte, CA, United States
| | - Mary Carroll
- Clinical Trial Office, City of Hope National Cancer Center, Duarte, CA, United States
| | - Vincent Chung
- Department of Medical Oncology & Therapeutics Research, City of Hope National Cancer Center, Duarte, CA, United States.
| |
Collapse
|
79
|
Wu RL, Sedlmeier G, Kyjacova L, Schmaus A, Philipp J, Thiele W, Garvalov BK, Sleeman JP. Hyaluronic acid-CD44 interactions promote BMP4/7-dependent Id1/3 expression in melanoma cells. Sci Rep 2018; 8:14913. [PMID: 30297743 PMCID: PMC6175841 DOI: 10.1038/s41598-018-33337-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/27/2018] [Indexed: 12/13/2022] Open
Abstract
BMP4/7-dependent expression of inhibitor of differentiation/DNA binding (Id) proteins 1 and 3 has been implicated in tumor progression and poor prognosis of malignant melanoma patients. Hyaluronic acid (HA), a pericellular matrix component, supports BMP7 signalling in murine chondrocytes through its receptor CD44. However, its role in regulating BMP signalling in melanoma is not clear. In this study we found that depletion of endogenously-produced HA by hyaluronidase treatment or by inhibition of HA synthesis by 4-methylumbelliferone (4-MU) resulted in reduced BMP4/7-dependent Id1/3 protein expression in mouse melanoma B16-F10 and Ret cells. Conversely, exogenous HA treatment increased BMP4/7-dependent Id1/3 protein expression. Knockdown of CD44 reduced BMP4/7-dependent Id1/3 protein expression, and attenuated the ability of exogenous HA to stimulate Id1 and Id3 expression in response to BMP. Co-IP experiments demonstrated that CD44 can physically associate with the BMP type II receptor (BMPR) ACVR2B. Importantly, we found that coordinate expression of Id1 or Id3 with HA synthases HAS2, HAS3, and CD44 is associated with reduced overall survival of cutaneous melanoma patients. Our results suggest that HA-CD44 interactions with BMPR promote BMP4/7-dependent Id1/3 protein expression in melanoma, contributing to reduced survival in melanoma patients.
Collapse
Affiliation(s)
- Ruo-Lin Wu
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany.,Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Georg Sedlmeier
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Lenka Kyjacova
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Anja Schmaus
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany.,KIT Campus Nord, Institute for Toxicology and Genetics, 76344, Karlsruhe, Germany
| | - Julia Philipp
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Wilko Thiele
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany.,KIT Campus Nord, Institute for Toxicology and Genetics, 76344, Karlsruhe, Germany
| | - Boyan K Garvalov
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Jonathan P Sleeman
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167, Mannheim, Germany. .,KIT Campus Nord, Institute for Toxicology and Genetics, 76344, Karlsruhe, Germany.
| |
Collapse
|
80
|
Contribution of Inhibitor of Differentiation and Estrogenic Endocrine Disruptors to Neurocognitive Disorders. Med Sci (Basel) 2018; 6:medsci6030061. [PMID: 30081481 PMCID: PMC6165108 DOI: 10.3390/medsci6030061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 01/17/2023] Open
Abstract
The devastating growth in the worldwide frequency of neurocognitive disorders and its allied difficulties, such as decline in memory, spatial competency, and ability to focus, poses a significant psychological public health problem. Inhibitor of differentiation (ID) proteins are members of a family of helix-loop-helix (HLH) transcription factors. ID proteins have been demonstrated to be involved in neurodevelopmental and depressive diseases and, thus, may influence neurocognitive deficiencies due to environmental exposure. Previously, it has been demonstrated that environmental factors, such as estrogenic endocrine disruptors (EEDs), have played an essential role in the influence of various neurocognitive disorders such as Alzheimer’s, dementia, and Parkinson’s disease. Based on this increasing number of reports, we consider the impact of these environmental pollutants on ID proteins. Better understanding of how these ID proteins by which EED exposure can affect neurocognitive disorders in populations will prospectively deliver valuable information in the impediment and regulation of these diseases linked with environmental factor exposure.
Collapse
|
81
|
Li X, Morrell NW, Yang J. Reply to "Letter to the Editor: Is Id3 proliferative or antiproliferative?". Am J Physiol Lung Cell Mol Physiol 2018; 315:L336-L337. [PMID: 30088800 DOI: 10.1152/ajplung.00315.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Science, Central South University , Changsha , China
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine , Cambridge , United Kingdom
| | - Jun Yang
- Department of Cell Biology, Institute of Basic Medical Sciences of Chinese Academy of Medical Sciences , Beijing , China
| |
Collapse
|
82
|
Vastrad C, Vastrad B. Bioinformatics analysis of gene expression profiles to diagnose crucial and novel genes in glioblastoma multiform. Pathol Res Pract 2018; 214:1395-1461. [PMID: 30097214 DOI: 10.1016/j.prp.2018.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/27/2018] [Accepted: 07/22/2018] [Indexed: 02/07/2023]
Abstract
Therefore, the current study aimed to diagnose the genes associated in the pathogenesis of GBM. The differentially expressed genes (DEGs) were diagnosed using the limma software package. The ToppFun was used to perform pathway and Gene Ontology (GO) enrichment analysis of the DEGs. Protein-protein interaction (PPI) networks, extracted modules, miRNA-target genes regulatory network and miRNA-target genes regulatory network were used to obtain insight into the actions of DEGs. Survival analysis for DEGs carried out. A total of 701 DEGs, including 413 upregulated and 288 downregulated genes, were diagnosed between U1118MG cell line (PK 11195 treated with 1 h exposure) and U1118MG cell line (PK 11195 treated with 24 h exposure). The up-regulated genes were enriched in superpathway of pyrimidine deoxyribonucleotides de novo biosynthesis, cell cycle, cell cycle process and chromosome. The down-regulated genes were enriched in folate transformations I, biosynthesis of amino acids, cellular amino acid metabolic process and vacuolar membrane. The current study screened the genes in PPI network, extracted modules, miRNA-target genes regulatory network and miRNA-target genes regulatory network with higher degrees as hub genes, which included MYC, TERF2IP, CDK1, EEF1G, TXNIP, SLC1A5, RGS4 and IER5L Survival suggested that low expressed NR4A2, SLC7 A5, CYR61 and ID1 in patients with GBM was linked with a positive prognosis for overall survival. In conclusion, the current study could improve our understanding of the molecular mechanisms in the progression of GBM, and these crucial as well as new molecular markers might be used as therapeutic targets for GBM.
Collapse
Affiliation(s)
- Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, 580001, Karanataka, India.
| | - Basavaraj Vastrad
- Department of Pharmaceutics, SET`S College of Pharmacy, Dharwad, Karnataka, 580002, India
| |
Collapse
|
83
|
Newman JH, Augeri DJ, NeMoyer R, Malhotra J, Langenfeld E, Chesson CB, Dobias NS, Lee MJ, Tarabichi S, Jhawar SR, Bommareddy PK, Marshall S, Sadimin ET, Kerrigan JE, Goedken M, Minerowicz C, Jabbour SK, Li S, Carayannopolous MO, Zloza A, Langenfeld J. Novel bone morphogenetic protein receptor inhibitor JL5 suppresses tumor cell survival signaling and induces regression of human lung cancer. Oncogene 2018; 37:3672-3685. [PMID: 29622797 PMCID: PMC10905627 DOI: 10.1038/s41388-018-0156-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 12/01/2017] [Accepted: 12/29/2017] [Indexed: 12/29/2022]
Abstract
BMP receptor inhibitors induce death of cancer cells through the downregulation of antiapoptotic proteins XIAP, pTAK1, and Id1-Id3. However, the current most potent BMP receptor inhibitor, DMH2, does not downregulate BMP signaling in vivo because of metabolic instability and poor pharmacokinetics. Here we identified the site of metabolic instability of DMH2 and designed a novel BMP receptor inhibitor, JL5. We show that JL5 has a greater volume of distribution and suppresses the expression of Id1 and pTak1 in tumor xenografts. Moreover, we demonstrate JL5-induced tumor cell death and tumor regression in xenograft mouse models without immune cells and humanized with adoptively transferred human immune cells. In humanized mice, JL5 additionally induces the infiltration of immune cells within the tumor microenvironment. Our studies show that the BMP signaling pathway is targetable in vivo and BMP receptor inhibitors can be developed as a therapeutic to treat cancer patients.
Collapse
Affiliation(s)
- Jenna H Newman
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - David J Augeri
- Office of Translational Science, Molecular Design and Synthesis, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Rachel NeMoyer
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Jyoti Malhotra
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Elaine Langenfeld
- Department of Surgery, Division of Surgical Oncology and Thoracic Surgery, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Charles B Chesson
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Natalie S Dobias
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Michael J Lee
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Saeed Tarabichi
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Sachin R Jhawar
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - Praveen K Bommareddy
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Sh'Rae Marshall
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Evita T Sadimin
- Department of Pathology and Laboratory Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - John E Kerrigan
- Department of Bioinformatics, Rutgers Biomedical Health Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Michael Goedken
- Office of Translational Science, Research Pathology Services, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Christine Minerowicz
- Department of Pathology and Laboratory Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - Shengguo Li
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Mary O Carayannopolous
- Department of Pathology and Laboratory Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - Andrew Zloza
- Section of Surgical Oncology Research, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08903, USA.
| | - John Langenfeld
- Department of Surgery, Division of Surgical Oncology and Thoracic Surgery, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
84
|
Row RH, Pegg A, Kinney BA, Farr GH, Maves L, Lowell S, Wilson V, Martin BL. BMP and FGF signaling interact to pattern mesoderm by controlling basic helix-loop-helix transcription factor activity. eLife 2018; 7:31018. [PMID: 29877796 PMCID: PMC6013256 DOI: 10.7554/elife.31018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 05/26/2018] [Indexed: 02/06/2023] Open
Abstract
The mesodermal germ layer is patterned into mediolateral subtypes by signaling factors including BMP and FGF. How these pathways are integrated to induce specific mediolateral cell fates is not well understood. We used mesoderm derived from post-gastrulation neuromesodermal progenitors (NMPs), which undergo a binary mediolateral patterning decision, as a simplified model to understand how FGF acts together with BMP to impart mediolateral fate. Using zebrafish and mouse NMPs, we identify an evolutionarily conserved mechanism of BMP and FGF-mediated mediolateral mesodermal patterning that occurs through modulation of basic helix-loop-helix (bHLH) transcription factor activity. BMP imparts lateral fate through induction of Id helix loop helix (HLH) proteins, which antagonize bHLH transcription factors, induced by FGF signaling, that specify medial fate. We extend our analysis of zebrafish development to show that bHLH activity is responsible for the mediolateral patterning of the entire mesodermal germ layer.
Collapse
Affiliation(s)
- Richard H Row
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Amy Pegg
- MRC Center for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Brian A Kinney
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Gist H Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, United States
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, United States.,Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, United States
| | - Sally Lowell
- MRC Center for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Valerie Wilson
- MRC Center for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| |
Collapse
|
85
|
Id2 Determines Intestinal Identity through Repression of the Foregut Transcription Factor Irx5. Mol Cell Biol 2018; 38:MCB.00250-17. [PMID: 29463648 PMCID: PMC5902590 DOI: 10.1128/mcb.00250-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 02/13/2018] [Indexed: 12/12/2022] Open
Abstract
The cellular components and function of the gastrointestinal epithelium exhibit distinct characteristics depending on the region, e.g., stomach or intestine. How these region-specific epithelial characteristics are generated during development remains poorly understood. Here, we report on the involvement of the helix-loop-helix inhibitor Id2 in establishing the specific characteristics of the intestinal epithelium. Id2−/− mice developed tumors in the small intestine. Histological analysis indicated that the intestinal tumors were derived from gastric metaplasia formed in the small intestine during development. Heterotopic Id2 expression in developing gastric epithelium induced a fate change to intestinal epithelium. Gene expression analysis revealed that foregut-enriched genes encoding Irx3 and Irx5 were highly induced in the midgut of Id2−/− embryos, and transgenic mice expressing Irx5 in the midgut endoderm developed tumors recapitulating the characteristics of Id2−/− mice. Altogether, our results demonstrate that Id2 plays a crucial role in the development of regional specificity in the gastrointestinal epithelium.
Collapse
|
86
|
Robichaux WG, Cheng X. Intracellular cAMP Sensor EPAC: Physiology, Pathophysiology, and Therapeutics Development. Physiol Rev 2018; 98:919-1053. [PMID: 29537337 PMCID: PMC6050347 DOI: 10.1152/physrev.00025.2017] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
This review focuses on one family of the known cAMP receptors, the exchange proteins directly activated by cAMP (EPACs), also known as the cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFs). Although EPAC proteins are fairly new additions to the growing list of cAMP effectors, and relatively "young" in the cAMP discovery timeline, the significance of an EPAC presence in different cell systems is extraordinary. The study of EPACs has considerably expanded the diversity and adaptive nature of cAMP signaling associated with numerous physiological and pathophysiological responses. This review comprehensively covers EPAC protein functions at the molecular, cellular, physiological, and pathophysiological levels; and in turn, the applications of employing EPAC-based biosensors as detection tools for dissecting cAMP signaling and the implications for targeting EPAC proteins for therapeutic development are also discussed.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| |
Collapse
|
87
|
Svendstrup M, Appel EVR, Sandholt CH, Ahluwalia TS, Ängquist LH, Thuesen BH, Jørgensen ME, Pedersen O, Grarup N, Hansen T, Sørensen TIA, Vestergaard H. Prospective Studies Exploring the Possible Impact of an ID3 Polymorphism on Changes in Obesity Measures. Obesity (Silver Spring) 2018; 26:747-754. [PMID: 29442437 DOI: 10.1002/oby.22109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/07/2017] [Accepted: 12/07/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Changes in fat mass depend on adipogenesis and angiogenesis, mechanisms regulated by the inhibitor of differentiation-3 (ID3). Id3 knockout mice showed attenuated increases in BMI and visceral fat mass. We hypothesized that the ID3 missense variant (rs11574-A) would lead to an attenuated increase over time in fat mass, BMI, waist circumference (WC), and waist-hip ratio (WHR) in humans. METHODS The genotyped study populations included the Obesity Research Group - Genetics (ORGGEN) cohort, a cohort of men with obesity (N = 716) and of randomly selected men (N = 826) from the Danish draft register who were examined at mean ages of 20 and 46 years, and the Inter99 (N = 6,116) and Health2006 (N = 2,761) cohorts, two population-based samples of middle-aged people, followed up after 5 years. RESULTS In meta-analyses of all data, no association was found between rs11574-A and changes in BMI, WC, WHR, or fat mass. We found an association between rs11574-A and cross-sectional BMI (N = 10,359, β: -0.16 kg/m2 per allele, 95% CI: -0.30 to -0.01, P = 0.033) and fat mass (N = 4,188, β: -0.52 kg/m2 per allele, 95% CI: -1.03 to -0.01, P = 0.046). CONCLUSIONS No consistent impact of the genetic variant on changes in fat mass, BMI, or fat distribution was found in three Danish cohorts.
Collapse
Affiliation(s)
- Mathilde Svendstrup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
- Danish Diabetes Academy, Odense, Denmark
| | - Emil V R Appel
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
| | - Camilla H Sandholt
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
| | - Tarunveer S Ahluwalia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Denmark
| | - Lars H Ängquist
- Department of Clinical Epidemiology, Bispebjerg and Frederiksberg Hospital, The Capital Region, Denmark
| | - Betina H Thuesen
- Research Center for Prevention and Health, Glostrup Hospital, Glostrup, Denmark
| | | | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Henrik Vestergaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Denmark
| |
Collapse
|
88
|
Ayob AZ, Ramasamy TS. Cancer stem cells as key drivers of tumour progression. J Biomed Sci 2018; 25:20. [PMID: 29506506 PMCID: PMC5838954 DOI: 10.1186/s12929-018-0426-4] [Citation(s) in RCA: 568] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/01/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are subpopulations of cancer cells sharing similar characteristics as normal stem or progenitor cells such as self-renewal ability and multi-lineage differentiation to drive tumour growth and heterogeneity. Throughout the cancer progression, CSC can further be induced from differentiated cancer cells via the adaptation and cross-talks with the tumour microenvironment as well as a response from therapeutic pressures, therefore contributes to their heterogeneous phenotypes. Challengingly, conventional cancer treatments target the bulk of the tumour and are unable to target CSCs due to their highly resistance nature, leading to metastasis and tumour recurrence. MAIN BODY This review highlights the roles of CSCs in tumour initiation, progression and metastasis with a focus on the cellular and molecular regulators that influence their phenotypical changes and behaviours in the different stages of cancer progression. We delineate the cross-talks between CSCs with the tumour microenvironment that support their intrinsic properties including survival, stemness, quiescence and their cellular and molecular adaptation in response to therapeutic pressure. An insight into the distinct roles of CSCs in promoting angiogenesis and metastasis has been captured based on in vitro and in vivo evidences. CONCLUSION Given dynamic cellular events along the cancer progression and contributions of resistance nature by CSCs, understanding their molecular and cellular regulatory mechanism in a heterogeneous nature, provides significant cornerstone for the development of CSC-specific therapeutics.
Collapse
Affiliation(s)
- Ain Zubaidah Ayob
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Wilayah Persekutuan Kuala Lumpur, Malaysia
- Cell and Molecular Laboratory (CMBL), The Dean’s Office, Faculty of Medicine, University of Malaya, 50603 Wilayah Persekutuan Kuala Lumpur, Malaysia
| |
Collapse
|
89
|
Aggarwal A, Feldman D, Feldman BJ. Identification of tumor-autonomous and indirect effects of vitamin D action that inhibit breast cancer growth and tumor progression. J Steroid Biochem Mol Biol 2018; 177:155-158. [PMID: 28710021 PMCID: PMC5764828 DOI: 10.1016/j.jsbmb.2017.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 12/19/2022]
Abstract
Several epidemiological studies have found that low vitamin D levels are associated with worse prognosis and poorer outcomes in patients with breast cancer (BCa), although some studies have failed to find this association. In addition, prior research has found that BCa patients with vitamin D deficiency have a more aggressive molecular phenotype and worse prognostic biomarkers. As vitamin D deficiency is common in patients diagnosed with BCa, elucidating the cause of the association between poor outcomes and vitamin D deficiency promises to have a significant impact on improving care for patients with BCa including enabling the development of novel therapeutic approaches. Here we review our recent findings in this area, including our data revealing that reduction of the expression of the vitamin D receptor (Vdr) within BCa cells accelerates primary tumor growth and enables the development of metastases, demonstrating a tumor autonomous effect of vitamin D signaling to suppress BCa metastases. We believe that these findings are likely relevant to humans as we discovered evidence that a mechanism of VDR regulation identified in our mouse models is conserved in human BCa. In particular, we identified a negative correlation between serum 25(OH)D concentration and the level of expression of the tumor progression factor ID1 in primary tumors from patients with breast cancer.
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pediatrics, Stanford School of Medicine, Stanford University, CA 94305, United States
| | - David Feldman
- Department of Medicine, Stanford School of Medicine, Stanford University, CA 94305, United States; Stanford Cancer Institute, Stanford University, CA 94305, United States
| | - Brian J Feldman
- Department of Pediatrics, Stanford School of Medicine, Stanford University, CA 94305, United States; Stanford Cancer Institute, Stanford University, CA 94305, United States.
| |
Collapse
|
90
|
Eguchi T, Sogawa C, Okusha Y, Uchibe K, Iinuma R, Ono K, Nakano K, Murakami J, Itoh M, Arai K, Fujiwara T, Namba Y, Murata Y, Ohyama K, Shimomura M, Okamura H, Takigawa M, Nakatsura T, Kozaki KI, Okamoto K, Calderwood SK. Organoids with cancer stem cell-like properties secrete exosomes and HSP90 in a 3D nanoenvironment. PLoS One 2018; 13:e0191109. [PMID: 29415026 PMCID: PMC5802492 DOI: 10.1371/journal.pone.0191109] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/28/2017] [Indexed: 12/12/2022] Open
Abstract
Ability to form cellular aggregations such as tumorspheres and spheroids have been used as a morphological marker of malignant cancer cells and in particular cancer stem cells (CSC). However, the common definition of the types of cellular aggregation formed by cancer cells has not been available. We examined morphologies of 67 cell lines cultured on three dimensional morphology enhancing NanoCulture Plates (NCP) and classified the types of cellular aggregates that form. Among the 67 cell lines, 49 cell lines formed spheres or spheroids, 8 cell lines formed grape-like aggregation (GLA), 8 cell lines formed other types of aggregation, and 3 cell lines formed monolayer sheets. Seven GLA-forming cell lines were derived from adenocarcinoma among the 8 lines. A neuroendocrine adenocarcinoma cell line PC-3 formed asymmetric GLA with ductal structures on the NCPs and rapidly growing asymmetric tumors that metastasized to lymph nodes in immunocompromised mice. In contrast, another adenocarcinoma cell line DU-145 formed spheroids in vitro and spheroid-like tumors in vivo that did not metastasize to lymph nodes until day 50 after transplantation. Culture in the 3D nanoenvironment and in a defined stem cell medium enabled the neuroendocrine adenocarcinoma cells to form slowly growing large organoids that expressed multiple stem cell markers, neuroendocrine markers, intercellular adhesion molecules, and oncogenes in vitro. In contrast, the more commonly used 2D serum-contained environment reduced intercellular adhesion and induced mesenchymal transition and promoted rapid growth of the cells. In addition, the 3D stemness nanoenvironment promoted secretion of HSP90 and EpCAM-exosomes, a marker of CSC phenotype, from the neuroendocrine organoids. These findings indicate that the NCP-based 3D environment enables cells to form stem cell tumoroids with multipotency and model more accurately the in vivo tumor status at the levels of morphology and gene expression.
Collapse
Affiliation(s)
- Takanori Eguchi
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School, Okayama, Japan
| | - Chiharu Sogawa
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuka Okusha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kenta Uchibe
- Department of Oral Morphology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - Kisho Ono
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Keisuke Nakano
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School, Okayama, Japan
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jun Murakami
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School, Okayama, Japan
- Department of Oral Diagnosis and Dent-maxillofacial Radiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Manabu Itoh
- JSR Life Sciences Corporation, Tsukuba, Japan
| | - Kazuya Arai
- JSR Life Sciences Corporation, Tsukuba, Japan
| | - Toshifumi Fujiwara
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuri Namba
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yoshiki Murata
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazumi Ohyama
- Radio Isotope Research Center, Okayama University Dental School, Okayama, Japan
| | - Manami Shimomura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hirohiko Okamura
- Department of Oral Morphology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School, Okayama, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Ken-ichi Kozaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Stuart K. Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
91
|
Lee JC, Kim IH, Cho JH, Lee TK, Park JH, Ahn JH, Shin BN, Yan BC, Kim JD, Jeon YH, Lee YJ, Won MH, Kang IJ. Vanillin improves scopolamine‑induced memory impairment through restoration of ID1 expression in the mouse hippocampus. Mol Med Rep 2018; 17:4399-4405. [PMID: 29328430 PMCID: PMC5802214 DOI: 10.3892/mmr.2018.8401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/26/2017] [Indexed: 11/06/2022] Open
Abstract
4-Hydroxy-3-methoxybenzaldehyde (vanillin), contained in a number of species of plant, has been reported to display beneficial effects against brain injuries. In the present study, the impact of vanillin on scopolamine‑induced alterations in cognition and the expression of DNA binding protein inhibitor ID‑1 (ID1), one of the inhibitors of DNA binding/differentiation proteins that regulate gene transcription, in the mouse hippocampus. Mice were treated with 1 mg/kg scopolamine with or without 40 mg/kg vanillin once daily for 4 weeks. Scopolamine‑induced cognitive impairment was observed from 1 week and was deemed to be severe 4 weeks following the administration of scopolamine. However, treatment with vanillin in scopolamine‑treated mice markedly attenuated cognitive impairment 4 weeks following treatment with scopolamine. ID1‑immunoreactive cells were revealed in the hippocampus of vehicle‑treated mice, and were hardly detected 4 weeks following treatment with scopolamine. However, treatment with vanillin in scopolamine‑treated mice markedly restored ID1‑immunoreactive cells and expression 4 weeks subsequent to treatment. The results of the present study suggested that vanillin may be beneficial for cognitive impairment, by preventing the reduction of ID1 expression which may be associated with cognitive impairment.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Bich Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Bing Chun Yan
- Institute of Integrative Traditional and Western Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jong-Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yong Hwan Jeon
- Department of Radiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Young Joo Lee
- Department of Emergency Medicine, Seoul Hospital, College of Medicine, Sooncheonhyang University, Seoul 04401, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| |
Collapse
|
92
|
Inhibitor of Differentiation-3 and Estrogenic Endocrine Disruptors: Implications for Susceptibility to Obesity and Metabolic Disorders. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6821601. [PMID: 29507860 PMCID: PMC5817379 DOI: 10.1155/2018/6821601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 11/07/2017] [Accepted: 11/23/2017] [Indexed: 12/28/2022]
Abstract
The rising global incidence of obesity cannot be fully explained within the context of traditional risk factors such as an unhealthy diet, physical inactivity, aging, or genetics. Adipose tissue is an endocrine as well as a metabolic organ that may be susceptible to disruption by environmental estrogenic chemicals. Since some of the endocrine disruptors are lipophilic chemicals with long half-lives, they tend to bioaccumulate in the adipose tissue of exposed populations. Elevated exposure to these chemicals may predispose susceptible individuals to weight gain by increasing the number and size of fat cells. Genetic studies have demonstrated that the transcriptional regulator inhibitor of differentiation-3 (ID3) promotes high fat diet-induced obesity in vivo. We have shown previously that PCB153 and natural estrogen 17β-estradiol increase ID3 expression. Based on our findings, we postulate that ID3 is a molecular target of estrogenic endocrine disruptors (EEDs) in the adipose tissue and a better understanding of this relationship may help to explain how EEDs can lead to the transcriptional programming of deviant fat cells. This review will discuss the current understanding of ID3 in excess fat accumulation and the potential for EEDs to influence susceptibility to obesity or metabolic disorders via ID3 signaling.
Collapse
|
93
|
Seong J, Kim NS, Kim JA, Lee W, Seo JY, Yum MK, Kim JH, Park I, Kang JS, Bae SH, Yun CH, Kong YY. Side branching and luminal lineage commitment by ID2 in developing mammary glands. Development 2018; 145:dev.165258. [DOI: 10.1242/dev.165258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022]
Abstract
Mammary glands develop through primary ductal elongation and side branching to maximize the spatial area. Although primary ducts are generated by bifurcation of terminal end buds, the mechanism through which side branching occurs is still largely unclear. Here, we show that inhibitor of DNA-binding 2 (ID2) drives side branch formation through differentiation of K6+ bipotent progenitor cells into CD61+ luminal progenitor cells. Id2-null mice had side branching defects, along with developmental blockage of K6+ bipotent progenitor cells into CD61+ luminal progenitor cells. Notably, CD61+ luminal progenitor cells were found in budding and side branches, but not in terminal end buds. Hormone reconstitution studies using ovariectomized MMTV-NLS-Id2 transgenic mice revealed that ID2 is a key mediator of progesterone, which drives luminal lineage differentiation and side branching. Our results suggest that CD61 is a marker for side branches and that ID2 regulates side branch formation by inducing luminal lineage commitment from K6+ bipotent progenitor cells to CD61+ luminal progenitor cells.
Collapse
Affiliation(s)
- Jinwoo Seong
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Nam-Shik Kim
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jee-Ah Kim
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Wonbin Lee
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ji-Yun Seo
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Min Kyu Yum
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ji-Hoon Kim
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Inkuk Park
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jong-Seol Kang
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Sung-Hwan Bae
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Young-Yun Kong
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| |
Collapse
|
94
|
Bolte C, Whitsett JA, Kalin TV, Kalinichenko VV. Transcription Factors Regulating Embryonic Development of Pulmonary Vasculature. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2018; 228:1-20. [PMID: 29288383 DOI: 10.1007/978-3-319-68483-3_1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lung morphogenesis is a highly orchestrated process beginning with the appearance of lung buds on approximately embryonic day 9.5 in the mouse. Endodermally derived epithelial cells of the primitive lung buds undergo branching morphogenesis to generate the tree-like network of epithelial-lined tubules. The pulmonary vasculature develops in close proximity to epithelial progenitor cells in a process that is regulated by interactions between the developing epithelium and underlying mesenchyme. Studies in transgenic and knockout mouse models demonstrate that normal lung morphogenesis requires coordinated interactions between cells lining the tubules, which end in peripheral saccules, juxtaposed to an extensive network of capillaries. Multiple growth factors, microRNAs, transcription factors, and their associated signaling cascades regulate cellular proliferation, migration, survival, and differentiation during formation of the peripheral lung. Dysregulation of signaling events caused by gene mutations, teratogens, or premature birth causes severe congenital and acquired lung diseases in which normal alveolar architecture and the pulmonary capillary network are disrupted. Herein, we review scientific progress regarding signaling and transcriptional mechanisms regulating the development of pulmonary vasculature during lung morphogenesis.
Collapse
Affiliation(s)
- Craig Bolte
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.,Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA. .,Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA. .,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
| |
Collapse
|
95
|
Bosch-Barrera J, Sais E, Cañete N, Marruecos J, Cuyàs E, Izquierdo A, Porta R, Haro M, Brunet J, Pedraza S, Menendez JA. Response of brain metastasis from lung cancer patients to an oral nutraceutical product containing silibinin. Oncotarget 2017; 7:32006-14. [PMID: 26959886 PMCID: PMC5077992 DOI: 10.18632/oncotarget.7900] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/21/2016] [Indexed: 12/31/2022] Open
Abstract
Despite multimodal treatment approaches, the prognosis of brain metastases (BM) from non-small cell lung cancer (NSCLC) remains poor. Untreated patients with BM have a median survival of about 1 month, with almost all patients dying from neurological causes. We herein present the first report describing the response of BM from NSCLC patients to an oral nutraceutical product containing silibinin, a flavonoid extracted from the seeds of the milk thistle. We present evidence of how the use of the silibinin-based nutraceutical Legasil® resulted in significant clinical and radiological improvement of BM from NSCLC patients with poor performance status that progressed after whole brain radiotherapy and chemotherapy. The suppressive effects of silibinin on progressive BM, which involved a marked reduction of the peritumoral brain edema, occurred without affecting the primary lung tumor outgrowth in NSCLC patients. Because BM patients have an impaired survival prognosis and are in need for an immediate tumor control, the combination of brain radiotherapy with silibinin-based nutraceuticals might not only alleviate BM edema but also prove local control and time for either classical chemotherapeutics with immunostimulatory effects or new immunotherapeutic agents such as checkpoint blockers to reveal their full therapeutic potential in NSCLC BM patients. New studies aimed to illuminate the mechanistic aspects underlying the regulatory effects of silibinin on the cellular and molecular pathobiology of BM might expedite the entry of new formulations of silibinin into clinical testing for progressive BM from lung cancer patients.
Collapse
Affiliation(s)
- Joaquim Bosch-Barrera
- Department of Medical Oncology, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital, Girona, Spain.,Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Elia Sais
- Department of Medical Oncology, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital, Girona, Spain
| | - Noemí Cañete
- Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Radiology, Diagnostic Imaging Institute, Doctor Josep Trueta University Hospital, Girona, Spain
| | - Jordi Marruecos
- Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Radiotherapy, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital, Girona, Spain
| | - Elisabet Cuyàs
- Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| | - Angel Izquierdo
- Department of Medical Oncology, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital, Girona, Spain.,Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Rut Porta
- Department of Medical Oncology, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital, Girona, Spain.,Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Manel Haro
- Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Medical Sciences, Medical School, University of Girona, Girona, Spain.,Department of Pneumology, Doctor Josep Trueta University Hospital, Girona, Spain
| | - Joan Brunet
- Department of Medical Oncology, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital, Girona, Spain.,Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Salvador Pedraza
- Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,Department of Medical Sciences, Medical School, University of Girona, Girona, Spain.,Department of Radiology, Diagnostic Imaging Institute, Doctor Josep Trueta University Hospital, Girona, Spain
| | - Javier A Menendez
- Girona Biomedical Research Institute (IDIBGi), Girona, Spain.,ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| |
Collapse
|
96
|
Roschger C, Verwanger T, Krammer B, Cabrele C. Reduction of cancer cell viability by synergistic combination of photodynamic treatment with the inhibition of the Id protein family. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 178:521-529. [PMID: 29245122 DOI: 10.1016/j.jphotobiol.2017.11.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/25/2017] [Accepted: 11/29/2017] [Indexed: 12/15/2022]
Abstract
The inhibitor of DNA binding and cell differentiation (Id) proteins are dominant negative regulators of the helix-loop-helix transcription factor family and play a key role during development as well as in vascular disorders and cancer. In fact, impairing the Id-protein activity in cancer cells reduces cell growth and even chemoresistance. Recently, we have shown that a synthetic Id-protein ligand (1Y) consisting of a cyclic nonapeptide can reduce the viability of the two breast cancer cell lines MCF-7 and T47D and of the bladder cancer cells T24 to about 50% at concentrations ≥100μM. Moreover, the cyclopeptide displays both proapoptotic and antiproliferative effects on MCF-7 cells. Herein, we show that the cyclopeptide does not induce cell death at the dose of 5μΜ, but it still inhibits MCF-7 and T24 cell proliferation, which correlates with an increased protein level of the cell-cycle regulator p27Kip1. Furthermore, 1Y-pretreated MCF-7, T47D, and T24 cells are more susceptible than untreated cells to the phototoxic effects of the three photosensitizers meta-tetra(hydroxyphenyl)chlorin, porfimer sodium, and hypericin, which are applied in photodynamic therapy (PDT). The combination of the Id-protein ligand with each of the light-activated photosensitizers shows synergistic effects on the reduction of cell viability. In conclusion, an Id-protein ligand with moderate cancer cell killing activity at concentrations ≥100μM can be applied at a 20-fold lower and barely toxic dose to raise the sensitivity of cancer cells towards phototoxicity associated with photodynamic treatment. This suggests the potential benefit of targeting the Id proteins in combined drug approaches for cancer therapy.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, 5020 Salzburg, Austria
| | - Thomas Verwanger
- Department of Molecular Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Barbara Krammer
- Department of Molecular Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, 5020 Salzburg, Austria.
| |
Collapse
|
97
|
Kepa A, Martinez Medina L, Erk S, Srivastava DP, Fernandes A, Toro R, Lévi S, Ruggeri B, Fernandes C, Degenhardt F, Witt SH, Meyer-Lindenberg A, Poncer JC, Martinot JL, Paillère Martinot ML, Müller CP, Heinz A, Walter H, Schumann G, Desrivières S. Associations of the Intellectual Disability Gene MYT1L with Helix-Loop-Helix Gene Expression, Hippocampus Volume and Hippocampus Activation During Memory Retrieval. Neuropsychopharmacology 2017; 42:2516-2526. [PMID: 28470180 PMCID: PMC5549840 DOI: 10.1038/npp.2017.91] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 12/27/2016] [Accepted: 01/17/2017] [Indexed: 01/06/2023]
Abstract
The fundamental role of the brain-specific myelin transcription factor 1-like (MYT1L) gene in cases of intellectual disability and in the etiology of neurodevelopmental disorders is increasingly recognized. Yet, its function remains under-investigated. Here, we identify a network of helix-loop-helix (HLH) transcriptional regulators controlled by MYT1L, as indicated by our analyses in human neural stem cells and in the human brain. Using cell-based knockdown approaches and microarray analyses we found that (1) MYT1L is required for neuronal differentiation and identified ID1, a HLH inhibitor of premature neurogenesis, as a target. (2) Although MYT1L prevented expression of ID1, it induced expression of a large number of terminal differentiation genes. (3) Consistently, expression of MYT1L in the human brain coincided with neuronal maturation and inversely correlated with that of ID1 and ID3 throughout the lifespan. (4) Genetic polymorphisms that reduced expression of MYT1L in the hippocampus resulted in increased expression of ID1 and ID3, decreased levels of the proneural basic HLH (bHLH) transcriptional regulators TCF4 and NEUROD6 and decreased expression of genes involved in long-term potentiation and synaptic transmission, cancer and neurodegeneration. Furthermore, our neuroimaging analyses indicated that MYT1L expression associated with hippocampal volume and activation during episodic memory recall, as measured by blood-oxygen-level-dependent (BOLD) signals. Overall, our findings suggest that MYT1L influences memory-related processes by controlling a neuronal proliferation/differentiation switch of ID-bHLH factors.
Collapse
Affiliation(s)
- Agnieszka Kepa
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Lourdes Martinez Medina
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Susanne Erk
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Deepak P Srivastava
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Alinda Fernandes
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Roberto Toro
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France,CNRS URA 2182, Genes, synapses and cognition, Institut Pasteur, Paris, France
| | - Sabine Lévi
- INSERM UMR-S 839, Paris, France,Université Pierre et Marie Curie, Paris, France,Institut du Fer a Moulin, Paris, France
| | - Barbara Ruggeri
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Cathy Fernandes
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Franziska Degenhardt
- Department of Genomics, Life and Brain Center, and Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | | | - Jean-Christophe Poncer
- INSERM UMR-S 839, Paris, France,Université Pierre et Marie Curie, Paris, France,Institut du Fer a Moulin, Paris, France
| | - Jean-Luc Martinot
- Institut National de la Sante et de la Recherche Medicale, INSERM CEAUnit1000, ‘‘Imaging & Psychiatry’’, IFR49, CEA, DSV, IBM-Service Hospitalier Frédéric Joliot, Orsay, France,University Paris Sud, Orsay, France,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Sante et de la Recherche Medicale, INSERM CEAUnit1000, ‘‘Imaging & Psychiatry’’, IFR49, CEA, DSV, IBM-Service Hospitalier Frédéric Joliot, Orsay, France,University Paris Sud, Orsay, France,Université Paris Descartes, Sorbonne Paris Cité, Paris, France,AP-HP Department of Adolescent Psychopathology and Medicine, Maison de Solenn, University Paris Descartes, Paris, France
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Gunter Schumann
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Sylvane Desrivières
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK,Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, Denmark Hill, London SE5 8AF, UK, Tel: +44(0)20 7848 0528, Fax: +44(0)20 7848 0866, E-mail:
| |
Collapse
|
98
|
Notch transactivates Rheb to maintain the multipotency of TSC-null cells. Nat Commun 2017; 8:1848. [PMID: 29184052 PMCID: PMC5705704 DOI: 10.1038/s41467-017-01845-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/20/2017] [Indexed: 02/07/2023] Open
Abstract
Differentiation abnormalities are a hallmark of tuberous sclerosis complex (TSC) manifestations; however, the genesis of these abnormalities remains unclear. Here we report on mechanisms controlling the multi-lineage, early neuronal progenitor and neural stem-like cell characteristics of lymphangioleiomyomatosis (LAM) and angiomyolipoma cells. These mechanisms include the activation of a previously unreported Rheb-Notch-Rheb regulatory loop, in which the cyclic binding of Notch1 to the Notch-responsive elements (NREs) on the Rheb promoter is a key event. This binding induces the transactivation of Rheb. The identified NRE2 and NRE3 on the Rheb promoter are important to Notch-dependent promoter activity. Notch cooperates with Rheb to block cell differentiation via similar mechanisms in mouse models of TSC. Cell-specific loss of Tsc1 within nestin-expressing cells in adult mice leads to the formation of kidney cysts, renal intraepithelial neoplasia, and invasive papillary renal carcinoma. Tuberous sclerosis complex (TSC) is a rare genetic condition causing tumours with differentiation abnormalities; however the molecular mechanisms causing these defects are unclear. Here the authors show that Notch cooperates with Rheb to block cell differentiation forming a regulatory loop that could underlie TSC tumorigenesis.
Collapse
|
99
|
Fu H, Hu D, Zhang L, Tang P. Role of extracellular vesicles in rheumatoid arthritis. Mol Immunol 2017; 93:125-132. [PMID: 29175592 DOI: 10.1016/j.molimm.2017.11.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 12/15/2022]
Abstract
Cell-derived extracellular vesicles (EVs) are involved in the pathogenesis of rheumatoid arthritis (RA), playing important roles in antigen presentation, inflammation, angiogenesis, cell-cell signal communication, thrombosis, and articular cartilage extracellular matrix degradation. Understanding the pathogenic mechanism of RA is important for developing therapies. The pathogenic indicators of RA, such as submicron-sized EVs, represent promising biomarkers for evaluating RA activity. This review summarizes the recent advances in understanding the pathogenesis of RA, and sheds light on the pathogenic as well as anti-inflammatory or immunosuppressive roles of EVs. We suggest that EVs could be harnessed as tools for drug delivery or targets for RA therapies.
Collapse
Affiliation(s)
- Haitao Fu
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300110, China
| | - Die Hu
- Qingdao Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Licheng Zhang
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Peifu Tang
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing 100853, China.
| |
Collapse
|
100
|
Gyoja F. Basic helix-loop-helix transcription factors in evolution: Roles in development of mesoderm and neural tissues. Genesis 2017; 55. [PMID: 28804953 DOI: 10.1002/dvg.23051] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/10/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022]
Abstract
Basic helix-loop-helix (bHLH) transcription factors have attracted the attention of developmental and evolutionary biologists for decades because of their conserved functions in mesodermal and neural tissue formation in both vertebrates and fruit flies. Their evolutionary history is of special interest because it will likely provide insights into developmental processes and refinement of metazoan-specific traits. This review briefly considers advances in developmental biological studies on bHLHs/HLHs. I also discuss recent genome-wide surveys and molecular phylogenetic analyses of these factors in a wide range of metazoans. I hypothesize that interactions between metazoan-specific Group A, D, and E bHLH/HLH factors enabled a sophisticated transition system from cell proliferation to differentiation in multicellular development. This control mechanism probably emerged initially to organize a multicellular animal body and was subsequently recruited to form evolutionarily novel tissues, which differentiated during a later ontogenetic phase.
Collapse
Affiliation(s)
- Fuki Gyoja
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan.,Department of Biology, Faculty of Science and Engineering, Konan University, 8-9-1 Okamoto, Higashinada, Kobe, 658-8501, Japan
| |
Collapse
|