1
|
Guo XF, Wang LL, Zheng FM, Li HP. PAX6 enhances Nanog expression by inhibiting NOTCH signaling to promote malignant properties in small cell lung cancer cells. Heliyon 2025; 11:e41795. [PMID: 39885876 PMCID: PMC11780942 DOI: 10.1016/j.heliyon.2025.e41795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/09/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive form of lung cancer. The aberrant regulation of Paired box 6 (PAX6) expression has been proposed to play an important oncogenic role in several cancer types. Nevertheless, there is limited knowledge regarding its function in SCLC. Here, we find that PAX6 overexpression promotes SCLC cell proliferation and cell cycle progression, while downregulation of PAX6 expression suppresses SCLC cell proliferation and cell cycle progression. Moreover, PAX6 enhances the enrichment of the ALDEFLUOR+ and CD133+ population and promotes sphere formation in SCLC cells. Additionally, upregulation or downregulation of PAX6 expression does not induce apoptosis in SCLC cells. Upregulation of PAX6 expression alleviates cisplatin or etoposide-induced apoptosis in SCLC cells, while downregulation of PAX6 expression aggravates cisplatin or etoposide-induced apoptosis in SCLC cells. Furthermore, PAX6 promotes the expression of stem cell factor Nanog. Interestingly, the downregulation of Nanog expression abolishes PAX6 promoted cell proliferation and cell cycle progression. Moreover, the inhibition of Nanog expression results in the inability of PAX6 to promote the increase of the ALDEFLUOR+ population, as well as the inability to mitigate apoptosis produced by cisplatin or etoposide in SCLC cells. Mechanically, PAX6 suppresses the activation of the NOTCH pathway to enhance Nanog expression. NOTCH pathway activation abolishes PAX6 promoted cell proliferation, cell cycle progression, ALDEFLUOR+ population enrichment, and apoptosis protection effect in SCLC cells. Our data indicates that PAX6 could be a critical factor for controlling cell proliferation, cell cycle progression, cancer stem cell properties, and apoptosis in SCLC.
Collapse
Affiliation(s)
- Xiao-Fang Guo
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ling-Ling Wang
- Department of Breast Surgery, Heyuan Women and Children's Hospital and Health Institute, Heyuan, Guangdong, China
| | - Fei-Meng Zheng
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - He-Ping Li
- Department of Medical Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Karami N, Taei A, Eftekhari-Yazdi P, Hassani F. Signaling pathway regulators in preimplantation embryos. J Mol Histol 2024; 56:57. [PMID: 39729177 DOI: 10.1007/s10735-024-10338-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Embryonic development during the preimplantation stages is highly sensitive and critically dependent on the reception of signaling cues. The precise coordination of diverse pathways and signaling factors is essential for successful embryonic progression. Even minor disruptions in these factors can result in physiological dysfunction, fetal malformations, or embryonic arrest. This issue is particularly evident in assisted reproductive technologies, such as in vitro fertilization, where embryonic arrest is frequently observed. A detailed understanding of these pathways enhances insight into the fundamental mechanisms underlying cellular processes and their contributions to embryonic development. The significance of elucidating signaling pathways and their regulatory factors in preimplantation development cannot be overstated. The application of this knowledge in laboratory settings has the potential to support strategies for modeling developmental stages and diseases, drug screening, therapeutic discovery, and reducing embryonic arrest. Furthermore, using various factors, small molecules, and pharmacological agents can enable the development or optimization of culture media for enhanced embryonic viability. While numerous pathways influence preimplantation development, this study examines several critical signaling pathways in this contex.
Collapse
Affiliation(s)
- Narges Karami
- MSc., Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O.Box 16635-148, Tehran, Iran
| | - Fatemeh Hassani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O.Box 16635-148, Tehran, Iran.
| |
Collapse
|
3
|
Shabbir S, Hadi A, Jabeen N, Hussain M. Developmental exposure of antibiotics shortens life span and induces teratogenicity in Drosophila melanogaster. Toxicol Rep 2024; 13:101784. [PMID: 39534686 PMCID: PMC11554921 DOI: 10.1016/j.toxrep.2024.101784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Antibiotics are the major therapeutic arsenal against bacterial infections. Yet, beneath this medical triumph lies an under investigated challenge of the potential teratological and toxicological impacts associated with the use of antibiotics. In the present study, we have explored the teratogenic potential of five commonly used antibiotics (streptomycin, metronidazole, tigecycline, doxycycline and norfloxacin) on Drosophila melanogaster Oregon-R strain. Except norfloxacin, all other tested antibiotics significantly delayed the onset of pupariation. Consistently, metronidazole, doxycycline and tigecycline resulted in statistically significant drops in egg-to-adult viability and adversely affected egg-to-pupa transition. In comparison, embryonic exposure of streptomycin impeded pupa-to-fly transition. All tested antibiotics induced morphological defects in antenna, wings, proboscis, eye, head, thorax, haltere and abdomen. Interestingly, developmental exposure of antibiotics resulted in statistically significant decrease in the lifespan of both male and female flies. This suggests an increased incidence of teratogenic faults at the systemic level, which are otherwise not manifested morphologically, due to the exposure of tested antibiotics during development. Taken together, our data show that developmental exposure of antibiotics may induce varying degrees of teratogenicity in D. melanogaster. Given the genomic homology and conservation of major molecular pathways that underpin development in humans and D. melanogaster, the findings do hold translational potential.
Collapse
Affiliation(s)
- Sanya Shabbir
- Bioinformatics and Molecular Medicine Research Group, Dow Fly Research Lab and Stock Centre, Dow College of Biotechnology, Dow University of Health Sciences, Karachi 75280, Pakistan
- Department of Microbiology, University of Karachi, Pakistan
| | - Abdullah Hadi
- Bioinformatics and Molecular Medicine Research Group, Dow Fly Research Lab and Stock Centre, Dow College of Biotechnology, Dow University of Health Sciences, Karachi 75280, Pakistan
| | - Nusrat Jabeen
- Department of Microbiology, University of Karachi, Pakistan
| | - Mushtaq Hussain
- Bioinformatics and Molecular Medicine Research Group, Dow Fly Research Lab and Stock Centre, Dow College of Biotechnology, Dow University of Health Sciences, Karachi 75280, Pakistan
| |
Collapse
|
4
|
Jarczak J, Bujko K, Ratajczak MZ, Kucia M. scRNA-seq revealed transcriptional signatures of human umbilical cord primitive stem cells and their germ lineage origin regulated by imprinted genes. Sci Rep 2024; 14:29264. [PMID: 39587190 PMCID: PMC11589151 DOI: 10.1038/s41598-024-79810-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
A population of CD133+lin-CD45- and CD34+lin-CD45- very small embryonic-like stem cells (VSELs) has been identified in postnatal human tissues, including bone marrow (BM), mobilized peripheral blood (mPB) and umbilical cord blood (UCB). Under appropriate conditions, VSELs in vitro and in vivo differentiate into tissue-committed stem cells for all three germ layers. Molecular analysis of adult murine BM-purified VSELs revealed that these rare cells deposited during development in adult tissues (i) express a similar transcriptome as embryonic stem cells, (ii) share several markers characteristic for epiblast and migratory primordial germ cells (PGCs), (iii) highly express a polycomb group protein enhancer of zeste drosophila homolog 2 (Ezh2) and finally (iv) display a unique pattern of imprinting at crucial paternally inherited genes that promotes their quiescence. Here, by employing single-cell RNA sequencing we demonstrate for the first time that purified from UCB human VSELs defined by expression of CD34 or CD133 antigens and lack of lineage markers, including CD45 antigen express similar molecular signature as murine BM-derived VSELs. Specifically, unsupervised clustering revealed numerous subpopulations of VSELs including ones i) annotated to germline compartments, ii) regulated by parental imprinting, iii) responding to early developmental fate decisions, iv) transcription factors involved in differentiation and development, including homeobox family of genes, and v) expressing innate immunity and purinergic signaling genes.
Collapse
Affiliation(s)
- Justyna Jarczak
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
| | - Kamila Bujko
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
| | - Mariusz Z Ratajczak
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
- Stem Cell Institute at Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Magdalena Kucia
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland.
| |
Collapse
|
5
|
Klinkovskij A, Shepelev M, Isaakyan Y, Aniskin D, Ulasov I. Advances of Genome Editing with CRISPR/Cas9 in Neurodegeneration: The Right Path towards Therapy. Biomedicines 2023; 11:3333. [PMID: 38137554 PMCID: PMC10741756 DOI: 10.3390/biomedicines11123333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
The rate of neurodegenerative disorders (NDDs) is rising rapidly as the world's population ages. Conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), and dementia are becoming more prevalent and are now the fourth leading cause of death, following heart disease, cancer, and stroke. Although modern diagnostic techniques for detecting NDDs are varied, scientists are continuously seeking new and improved methods to enable early and precise detection. In addition to that, the present treatment options are limited to symptomatic therapy, which is effective in reducing the progression of neurodegeneration but lacks the ability to target the root cause-progressive loss of neuronal functioning. As a result, medical researchers continue to explore new treatments for these conditions. Here, we present a comprehensive summary of the key features of NDDs and an overview of the underlying mechanisms of neuroimmune dysfunction. Additionally, we dive into the cutting-edge treatment options that gene therapy provides in the quest to treat these disorders.
Collapse
Affiliation(s)
- Aleksandr Klinkovskij
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia; (A.K.); (D.A.)
| | - Mikhail Shepelev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Str., Moscow 119334, Russia
| | - Yuri Isaakyan
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8 Trubetskaya Str., Moscow 119991, Russia;
| | - Denis Aniskin
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia; (A.K.); (D.A.)
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia; (A.K.); (D.A.)
| |
Collapse
|
6
|
Martinez Lyons A, Boulter L. NOTCH signalling - a core regulator of bile duct disease? Dis Model Mech 2023; 16:dmm050231. [PMID: 37605966 PMCID: PMC10461466 DOI: 10.1242/dmm.050231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023] Open
Abstract
The Notch signalling pathway is an evolutionarily conserved mechanism of cell-cell communication that mediates cellular proliferation, fate determination and maintenance of stem/progenitor cell populations across tissues. Although it was originally identified as a critical regulator of embryonic liver development, NOTCH signalling activation has been associated with the pathogenesis of a number of paediatric and adult liver diseases. It remains unclear, however, what role NOTCH actually plays in these pathophysiological processes and whether NOTCH activity represents the reactivation of a conserved developmental programme that is essential for adult tissue repair. In this Review, we explore the concepts that NOTCH signalling reactivation in the biliary epithelium is a reiterative and essential response to bile duct damage and that, in disease contexts in which biliary epithelial cells need to be regenerated, NOTCH signalling supports ductular regrowth. Furthermore, we evaluate the recent literature on NOTCH signalling as a critical factor in progenitor-mediated hepatocyte regeneration, which indicates that the mitogenic role for NOTCH signalling in biliary epithelial cell proliferation has also been co-opted to support other forms of epithelial regeneration in the adult liver.
Collapse
Affiliation(s)
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
- CRUK Scottish Centre, Institute of Genetics and Cancer, Edinburgh EH4 2XU, UK
| |
Collapse
|
7
|
Jiang Z, Wu F, Laise P, Takayuki T, Na F, Kim W, Kobayashi H, Chang W, Takahashi R, Valenti G, Sunagawa M, White RA, Macchini M, Renz BW, Middelhoff M, Hayakawa Y, Dubeykovskaya ZA, Tan X, Chu TH, Nagar K, Tailor Y, Belin BR, Anand A, Asfaha S, Finlayson MO, Iuga AC, Califano A, Wang TC. Tff2 defines transit-amplifying pancreatic acinar progenitors that lack regenerative potential and are protective against Kras-driven carcinogenesis. Cell Stem Cell 2023; 30:1091-1109.e7. [PMID: 37541213 PMCID: PMC10414754 DOI: 10.1016/j.stem.2023.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 04/06/2023] [Accepted: 07/07/2023] [Indexed: 08/06/2023]
Abstract
While adult pancreatic stem cells are thought not to exist, it is now appreciated that the acinar compartment harbors progenitors, including tissue-repairing facultative progenitors (FPs). Here, we study a pancreatic acinar population marked by trefoil factor 2 (Tff2) expression. Long-term lineage tracing and single-cell RNA sequencing (scRNA-seq) analysis of Tff2-DTR-CreERT2-targeted cells defines a transit-amplifying progenitor (TAP) population that contributes to normal homeostasis. Following acute and chronic injury, Tff2+ cells, distinct from FPs, undergo depopulation but are eventually replenished. At baseline, oncogenic KrasG12D-targeted Tff2+ cells are resistant to PDAC initiation. However, KrasG12D activation in Tff2+ cells leads to survival and clonal expansion following pancreatitis and a cancer stem/progenitor cell-like state. Selective ablation of Tff2+ cells prior to KrasG12D activation in Mist1+ acinar or Dclk1+ FP cells results in enhanced tumorigenesis, which can be partially rescued by adenoviral Tff2 treatment. Together, Tff2 defines a pancreatic TAP population that protects against Kras-driven carcinogenesis.
Collapse
Affiliation(s)
- Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Feijing Wu
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Pasquale Laise
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA; DarwinHealth Inc., New York, NY, USA
| | - Tanaka Takayuki
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Fu Na
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Woosook Kim
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Hiroki Kobayashi
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Wenju Chang
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Ryota Takahashi
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Giovanni Valenti
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Masaki Sunagawa
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Ruth A White
- Division of Hematology and Oncology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Marina Macchini
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Bernhard W Renz
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of General, Visceral, and Transplantation Surgery, LMU University Hospital, LMU Munich, Germany
| | - Moritz Middelhoff
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Division of Digestive and Liver Diseases, CU and Klinikum rechts der Isar, Technical University, Munich, Germany
| | - Yoku Hayakawa
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Zinaida A Dubeykovskaya
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Xiangtian Tan
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Timothy H Chu
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Karan Nagar
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Yagnesh Tailor
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Bryana R Belin
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Akanksha Anand
- Division of Digestive and Liver Diseases, Department of Medicine and Department of Gastroenterology II, Klinikum rechts der Isar, Technical University, Munich, Germany
| | - Samuel Asfaha
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Michael O Finlayson
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Alina C Iuga
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA; DarwinHealth Inc., New York, NY, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
8
|
Tian Y, Zhang P, Mou Y, Yang W, Zhang J, Li Q, Dou X. Silencing Notch4 promotes tumorigenesis and inhibits metastasis of triple-negative breast cancer via Nanog and Cdc42. Cell Death Discov 2023; 9:148. [PMID: 37149651 PMCID: PMC10164131 DOI: 10.1038/s41420-023-01450-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023] Open
Abstract
Elucidation of individual Notch protein biology in specific cancer is crucial to develop safe, effective, and tumor-selective Notch-targeting therapeutic reagents for clinical use [1]. Here, we explored the Notch4 function in triple-negative breast cancer (TNBC). We found that silencing Notch4 enhanced tumorigenic ability in TNBC cells via upregulating Nanog expression, a pluripotency factor of embryonic stem cells. Intriguingly, silencing Notch4 in TNBC cells suppressed metastasis via downregulating Cdc42 expression, a key molecular for cell polarity formation. Notably, downregulation of Cdc42 expression affected Vimentin distribution, but not Vimentin expression to inhibit EMT shift. Collectively, our results show that silencing Notch4 enhances tumorigenesis and inhibits metastasis in TNBC, indicating that targeting Notch4 may not be a potential strategy for drug discovery in TNBC.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Peipei Zhang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Yajun Mou
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Wenxiu Yang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Junhong Zhang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Qing Li
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China
| | - Xiaowei Dou
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, China.
| |
Collapse
|
9
|
Huynh K, Smith BR, Macdonald SJ, Long AD. Genetic variation in chromatin state across multiple tissues in Drosophila melanogaster. PLoS Genet 2023; 19:e1010439. [PMID: 37146087 PMCID: PMC10191298 DOI: 10.1371/journal.pgen.1010439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 05/17/2023] [Accepted: 04/20/2023] [Indexed: 05/07/2023] Open
Abstract
We use ATAC-seq to examine chromatin accessibility for four different tissues in Drosophila melanogaster: adult female brain, ovaries, and both wing and eye-antennal imaginal discs from males. Each tissue is assayed in eight different inbred strain genetic backgrounds, seven associated with a reference quality genome assembly. We develop a method for the quantile normalization of ATAC-seq fragments and test for differences in coverage among genotypes, tissues, and their interaction at 44099 peaks throughout the euchromatic genome. For the strains with reference quality genome assemblies, we correct ATAC-seq profiles for read mis-mapping due to nearby polymorphic structural variants (SVs). Comparing coverage among genotypes without accounting for SVs results in a highly elevated rate (55%) of identifying false positive differences in chromatin state between genotypes. After SV correction, we identify 1050, 30383, and 4508 regions whose peak heights are polymorphic among genotypes, among tissues, or exhibit genotype-by-tissue interactions, respectively. Finally, we identify 3988 candidate causative variants that explain at least 80% of the variance in chromatin state at nearby ATAC-seq peaks.
Collapse
Affiliation(s)
- Khoi Huynh
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California, United States of America
| | - Brittny R. Smith
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Stuart J. Macdonald
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, United States of America
| | - Anthony D. Long
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California, United States of America
| |
Collapse
|
10
|
Gao Z, Ni X, Zheng B, Sun W, Wan W, Liu H, Ni X, Suo T, Li N, Liu H, Shen S. Biliverdin reductase B impairs cholangiocarcinoma cell motility by inhibiting the Notch/Snail signaling pathway. J Cancer 2022; 13:2159-2170. [PMID: 35517415 PMCID: PMC9066219 DOI: 10.7150/jca.70323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/22/2022] [Indexed: 12/04/2022] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most lethal types of solid tumors worldwide. Lymph node metastasis is common in the early stage, which is associated with recurrence and reduced survival time after CCA resection. The molecular pathogenesis of CCA is complex and requires extensive investigation. It involves multiple genomic alterations and the dysregulation of signaling pathways. Biliverdin reductase B (BLVRB) is a non-redundant NAD(P)H-dependent biliverdin reductase that regulates cellular redox status by reducing biliverdin to bilirubin. This study aimed at describing the biological functions and molecular mechanisms of BLVRB in human CCA. Prognostic clinical data showed that low expression BLVRB was associated with poor prognosis and lymph node metastasis. BLVRB depletion accelerated epithelial-mesenchymal transition (EMT), cell migration and invasion. In contrast, BLVRB overexpression was associated with reduced EMT and cell migration and invasion in CCA. BLVRB suppression activated Notch signaling, and activated c-Notch enhanced EMT by upregulating Snail expression levels, thereby increasing cell migration and invasion in CCA. Our results identified an unexpected function of BLVRB in CCA migration and invasion through the regulation of Notch/Snail signaling.
Collapse
Affiliation(s)
- Zhihui Gao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Xiaojian Ni
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Bohao Zheng
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Wentao Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Wenze Wan
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Han Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Xiaoling Ni
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Tao Suo
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Na Li
- Basic Medical Institute; Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Houbao Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
- Department of General Surgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, China
| | - Sheng Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
- Department of General Surgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, China
| |
Collapse
|
11
|
Holtzer L, Wesseling-Rozendaal Y, Verhaegh W, van de Stolpe A. Measurement of activity of developmental signal transduction pathways to quantify stem cell pluripotency and phenotypically characterize differentiated cells. Stem Cell Res 2022; 61:102748. [PMID: 35325817 DOI: 10.1016/j.scr.2022.102748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/28/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022] Open
Abstract
Important challenges in stem cell research and regenerative medicine are reliable assessment of pluripotency state and purity of differentiated cell populations. Pluripotency and differentiation are regulated and determined by activity of developmental signal transduction pathways (STPs). To date activity of these STPs could not be directly measured on a cell sample. Here we validate a novel assay platform for measurement of activity of developmental STPs (STP) for use in stem cells and stem cell derivatives. In addition to previously developed STP assays, we report development of an additional STP assay for the MAPK-AP1 pathway. Subsequently, activity of Notch, Hedgehog, TGFβ, Wnt, PI3K, MAPK-AP1, and NFκB signaling pathways was calculated from Affymetrix transcriptome data of human pluripotent embryonic (hES) and iPS cell lines under different culture conditions, organ-derived multipotent stem cells, and differentiated cell types, to generate quantitative STP activity profiles. Results show that the STP assay technology enables reliable and quantitative measurement of multiple STP activities simultaneously on any individual cell sample. Using the technology, we found that culture conditions dominantly influence the pluripotent stem cell STP activity profile, while the origin of the stem cell line was a minor variable. A pluripotency STP activity profile (Pluripotency qPAP) was defined (active PI3K, MAPK, Hedgehog, Notch, TGFβ, and NFκB pathway, inactive Wnt pathway). Differentiation of hES cells to intestinal progenitor cells resulted in an STP activity profile characterized by active PI3K, Wnt and Notch pathways, comparable to the STP activity profile measured on primary intestinal crypt stem cells. Quantitative STP activity measurement is expected to improve experimental reproducibility and standardization of pluripotent and multipotent stem cell culture/differentiation, and enable controlled manipulation of pluripotency/differentiation state using pathway targeting compounds.
Collapse
Affiliation(s)
- Laurent Holtzer
- Molecular Pathway Diagnostics, Philips, Eindhoven, The Netherlands.
| | | | - Wim Verhaegh
- Molecular Pathway Diagnostics, Philips, Eindhoven, The Netherlands.
| | | |
Collapse
|
12
|
Dietrich B, Haider S, Meinhardt G, Pollheimer J, Knöfler M. WNT and NOTCH signaling in human trophoblast development and differentiation. Cell Mol Life Sci 2022; 79:292. [PMID: 35562545 PMCID: PMC9106601 DOI: 10.1007/s00018-022-04285-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/25/2022] [Accepted: 04/01/2022] [Indexed: 12/16/2022]
Abstract
Correct development of the human placenta and its differentiated epithelial cells, syncytial trophoblasts (STBs) and extravillous trophoblasts (EVTs), is crucial for a successful pregnancy outcome. STBs develop by cell fusion of mononuclear cytotrophoblasts (CTBs) in placental floating villi, whereas migratory EVTs originate from specialized villi anchoring to the maternal decidua. Defects in trophoblast differentiation have been associated with severe pregnancy disorders such as early-onset preeclampsia and fetal growth restriction. However, the evolutionary pathways underlying normal and adverse placentation are poorly understood. Herein, we discuss Wingless (WNT) and NOTCH signaling, two pathways that play pivotal roles in human placenta and trophoblast development. Whereas WNT is necessary for expansion of trophoblast progenitors and stem cells, NOTCH1 is required for proliferation and survival of EVT precursors. Differentiation of the latter is orchestrated by a switch in NOTCH receptor expression as well as by changes in WNT ligands and their downstream effectors.
Collapse
Affiliation(s)
- Bianca Dietrich
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Sandra Haider
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Gudrun Meinhardt
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Jürgen Pollheimer
- grid.22937.3d0000 0000 9259 8492Maternal-Fetal Immunology Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Martin Knöfler
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| |
Collapse
|
13
|
Muhr J, Hagey DW. The cell cycle and differentiation as integrated processes: Cyclins and CDKs reciprocally regulate Sox and Notch to balance stem cell maintenance. Bioessays 2021; 43:e2000285. [PMID: 34008221 DOI: 10.1002/bies.202000285] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022]
Abstract
Development and maintenance of diverse organ systems require context-specific regulation of stem cell behaviour. We hypothesize that this is achieved via reciprocal regulation between the cell cycle machinery and differentiation factors. This idea is supported by the parallel evolutionary emergence of differentiation pathways, cell cycle components and complex multicellularity. In addition, the activities of different cell cycle phases have been found to bias cells towards stem cell maintenance or differentiation. Finally, several direct mechanistic links between these two processes have been established. Here, we focus on interactions between cyclin-CDK complexes and differentiation regulators of the Notch pathway and Sox family of transcription factors within the context of pluripotent and neural stem cells. Thus, this hypothesis formalizes the links between these two processes as an integrated network. Since such factors are common to all stem cells, better understanding their interconnections will help to explain their behaviour in health and disease.
Collapse
Affiliation(s)
- Jonas Muhr
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Daniel W Hagey
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
14
|
Ghosh A, Som A. Decoding molecular markers and transcriptional circuitry of naive and primed states of human pluripotency. Stem Cell Res 2021; 53:102334. [PMID: 33862536 DOI: 10.1016/j.scr.2021.102334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 11/17/2022] Open
Abstract
Pluripotent stem cells (PSCs) have been observed to occur in two distinct states - naive and primed. Both naive and primed state PSCs can give rise to tissues of all the three germ layers in vitro but differ in their potential to generate germline chimera in vivo. Understanding the molecular mechanisms that govern these two states of pluripotency in human can open a plethora of opportunities for studying early embryonic development and in biomedical applications. In this work, we use weighted gene co-expression network analysis (WGCNA) to identify the key molecular makers and their interactions that define the two distinct pluripotency states. Signed hybrid network was reconstructed from transcriptomic data (RNA-seq) of naive and primed state pluripotent samples. Our analysis revealed two sets of genes that are involved in the establishment and maintenance of naive and primed states. The naive state genes were found to be enriched for biological processes and pathways related to metabolic processes while primed state genes were associated with system development. We further filtered these lists to identify the intra-modular hubs and the hub transcription factors (TFs) for each group. Validation of the identified TFs was carried out using independent microarray datasets and we finally present a list of 52 and 33 TFs as the set of core TFs that are responsible for the induction and maintenance of naive and primed states of pluripotency in human, respectively. Among these, the TFs ZNF275, ZNF232, SP4, and MSANTD3 could be of interest as they were not reported in previous studies.
Collapse
Affiliation(s)
- Arindam Ghosh
- Centre of Bioinformatics, Institute of Interdisciplinary Studies, University of Allahabad, Prayagraj 211002, India; Institute of Biomedicine, University of Eastern Finland, FI-70210 Kuopio, Finland
| | - Anup Som
- Centre of Bioinformatics, Institute of Interdisciplinary Studies, University of Allahabad, Prayagraj 211002, India.
| |
Collapse
|
15
|
Crosstalk between miRNAs and signaling pathways involved in pancreatic cancer and pancreatic ductal adenocarcinoma. Eur J Pharmacol 2021; 901:174006. [PMID: 33711308 DOI: 10.1016/j.ejphar.2021.174006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/19/2021] [Accepted: 03/02/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer (PC) is the seventh leading cause of cancer-related deaths worldwide with 5-year survival rates below 8%. Most patients with PC and pancreatic ductal adenocarcinoma (PDAC) die after relapse and cancer progression as well as resistance to treatment. Pancreatic tumors contain a high desmoplastic stroma that forms a rigid mass and has a potential role in tumor growth and metastasis. PC initiates from intraepithelial neoplasia lesions leading to invasive cancer through various pathways. These lesions harbor particular changes in signaling pathways involved in the tumorigenesis process. These events affect both the epithelial cells, including the tumor and the surrounding stroma, and eventually lead to the formation of complex signaling networks. Genetic studies of PC have revealed common molecular features such as the presence of mutations in KRAS gene in more than 90% of patients, as well as the inactivation or deletion mutations of some tumor suppressor genes including TP53, CDKN2A, and SMAD4. In recent years, studies have also identified different roles of microRNAs in PC pathogenesis as well as their importance in PC diagnosis and treatment, and their involvement in various signaling pathways. In this study, we discussed the most common pathways involved in PC and PDAC as well as their role in tumorigenesis and progression. Furthermore, the miRNAs participating in the regulation of these signaling pathways in PC progression are summarized in this study. Therefore, understanding more about pathways involved in PC can help with the development of new and effective therapies in the future.
Collapse
|
16
|
Lee J, Cho Y. Potential roles of stem cell marker genes in axon regeneration. Exp Mol Med 2021; 53:1-7. [PMID: 33446881 PMCID: PMC8080715 DOI: 10.1038/s12276-020-00553-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/16/2020] [Indexed: 01/29/2023] Open
Abstract
Axon regeneration is orchestrated by many genes that are differentially expressed in response to injury. Through a comparative analysis of gene expression profiling, injury-responsive genes that are potential targets for understanding the mechanisms underlying regeneration have been revealed. As the efficiency of axon regeneration in both the peripheral and central nervous systems can be manipulated, we suggest that identifying regeneration-associated genes is a promising approach for developing therapeutic applications in vivo. Here, we review the possible roles of stem cell marker- or stemness-related genes in axon regeneration to gain a better understanding of the regeneration mechanism and to identify targets that can enhance regenerative capacity.
Collapse
Affiliation(s)
- Jinyoung Lee
- Laboratory of Axon Regeneration & Degeneration, Department of Life Sciences, Korea University, Anam-ro 145, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Yongcheol Cho
- Laboratory of Axon Regeneration & Degeneration, Department of Life Sciences, Korea University, Anam-ro 145, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| |
Collapse
|
17
|
Papadopoulos A, Chalmantzi V, Mikhaylichenko O, Hyvönen M, Stellas D, Kanhere A, Heath J, Cunningham DL, Fotsis T, Murphy C. Combined transcriptomic and phosphoproteomic analysis of BMP4 signaling in human embryonic stem cells. Stem Cell Res 2020; 50:102133. [PMID: 33383406 DOI: 10.1016/j.scr.2020.102133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 11/08/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022] Open
Abstract
Human embryonic stem cells (hESCs) are an invaluable tool in the fields of embryology and regenerative medicine. Activin A and BMP4 are well-characterised growth factors implicated in pluripotency and differentiation. In the current study, hESCs are cultured in a modified version of mTeSR1, where low concentrations of ActivinA substitute for TGFβ. This culture system is further used to investigate the changes induced by BMP4 on hESCs by employing a combination of transcriptomic and phosphoproteomic approaches. Results indicate that in a pluripotent state, hESCs maintain WNT signaling under negative regulation by expressing pathway inhibitors. Initial stages of differentiation are characterized by upregulation of WNT pathway ligands, TGFβ pathway inhibitors which have been shown in Xenopus to expand the BMP signaling range essential for embryonic patterning, and mesendodermal transcripts. Moreover, BMP4 enhances the phosphorylation of proteins associated with migration and transcriptional regulation. Results further indicate the vital regulatory role of Activin A and BMP4 in crucial fate decisions in hESCs.
Collapse
Affiliation(s)
- Angelos Papadopoulos
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, United Kingdom
| | - Varvara Chalmantzi
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Olga Mikhaylichenko
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, United Kingdom
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, United Kingdom
| | - Dimitris Stellas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Aditi Kanhere
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - John Heath
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Debbie L Cunningham
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Theodore Fotsis
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, University Campus of Ioannina, 45110 Ioannina, Greece; Laboratory of Biology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Carol Murphy
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, University Campus of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
18
|
Zohorsky K, Mequanint K. Designing Biomaterials to Modulate Notch Signaling in Tissue Engineering and Regenerative Medicine. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:383-410. [PMID: 33040694 DOI: 10.1089/ten.teb.2020.0182] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The design of cell-instructive biomaterials for tissue engineering and regenerative medicine is at a crossroads. Although the conventional tissue engineering approach is top-down (cells seeded to macroporous scaffolds and mature to form tissues), bottom-up tissue engineering strategies are becoming appealing. With such developments, we can study cell signaling events, thus enabling functional tissue assembly in physiologic and diseased models. Among many important signaling pathways, the Notch signaling pathway is the most diverse in its influence during tissue morphogenesis and repair following injury. Although Notch signaling is extensively studied in developmental biology and cancer biology, our knowledge of designing biomaterial-based Notch signaling platforms and incorporating Notch signaling components into engineered tissue systems is limited. By incorporating Notch signaling to tissue engineering scaffolds, we can direct cell-specific responses and improve engineered tissue maturation. This review will discuss recent progress in the development of Notch signaling biomaterials as a promising target to control cellular fate decisions, including the influences of ligand identity, biophysical material cues, ligand presentation strategies, and mechanotransduction. Notch signaling is consequently of interest to direct, control, and reprogram cellular behavior on a biomaterial surface. We anticipate that discussions in this article will allow for enhanced knowledge and insight into designing Notch targeted biomaterials for various tissue engineering and cell fate determinations. Impact statement Notch signaling is recognized as an important pathway in tissue engineering and regenerative medicine; however, there is no systematic review on this topic. The comprehensive review and perspectives presented here provide an in-depth discussion on ligand presentation strategies both in 2D and in 3D cell culture environments involving biomaterials/scaffolds. In addition, this review article provides insight into the challenges in designing cell surrogate biomaterials capable of providing Notch signals. To the best of the authors' knowledge, this is the first review relevant to the fields of tissue engineering.
Collapse
Affiliation(s)
- Kathleen Zohorsky
- School of Biomedical Engineering and The University of Western Ontario, London, Canada
| | - Kibret Mequanint
- School of Biomedical Engineering and The University of Western Ontario, London, Canada.,Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Canada
| |
Collapse
|
19
|
Wuputra K, Ku CC, Wu DC, Lin YC, Saito S, Yokoyama KK. Prevention of tumor risk associated with the reprogramming of human pluripotent stem cells. J Exp Clin Cancer Res 2020; 39:100. [PMID: 32493501 PMCID: PMC7268627 DOI: 10.1186/s13046-020-01584-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023] Open
Abstract
Human pluripotent embryonic stem cells have two special features: self-renewal and pluripotency. It is important to understand the properties of pluripotent stem cells and reprogrammed stem cells. One of the major problems is the risk of reprogrammed stem cells developing into tumors. To understand the process of differentiation through which stem cells develop into cancer cells, investigators have attempted to identify the key factors that generate tumors in humans. The most effective method for the prevention of tumorigenesis is the exclusion of cancer cells during cell reprogramming. The risk of cancer formation is dependent on mutations of oncogenes and tumor suppressor genes during the conversion of stem cells to cancer cells and on the environmental effects of pluripotent stem cells. Dissecting the processes of epigenetic regulation and chromatin regulation may be helpful for achieving correct cell reprogramming without inducing tumor formation and for developing new drugs for cancer treatment. This review focuses on the risk of tumor formation by human pluripotent stem cells, and on the possible treatment options if it occurs. Potential new techniques that target epigenetic processes and chromatin regulation provide opportunities for human cancer modeling and clinical applications of regenerative medicine.
Collapse
Affiliation(s)
- Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 807, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 807, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Deng-Chyang Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Ying-Chu Lin
- School of Dentistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Shigeo Saito
- Waseda University Research Institute for Science and Engineering, Shinjuku, Tokyo, 162-8480, Japan.
- Saito Laboratory of Cell Technology Institute, Yaita, Tochigi, 329-1571, Japan.
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 807, Taiwan.
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Waseda University Research Institute for Science and Engineering, Shinjuku, Tokyo, 162-8480, Japan.
| |
Collapse
|
20
|
Atypical chemokine receptor ACKR3/CXCR7 controls postnatal vasculogenesis and arterial specification by mesenchymal stem cells via Notch signaling. Cell Death Dis 2020; 11:307. [PMID: 32366833 PMCID: PMC7198625 DOI: 10.1038/s41419-020-2512-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are known to play a role in postnatal vasculogenesis and hold great promise for vascular regeneration. However, the mechanisms by which the endothelial differentiation and specification of MSCs remain unclear. We examined the potential role and molecular mechanisms of atypical chemokine receptor ACKR3/CXCR7 in MSC-mediated endothelial cell differentiation and specification. Here, we showed that vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) activate CXCR7 expression on MSCs through PDGF receptors, PDGFRα and PDGFRβ-mediated phosphoinositide 3-kinase (PI3K)/Akt signaling. Genetic and pharmacologic blockage of CXCR7 on MSCs suppressed the VEGF or stromal cell-derived factor 1 (SDF)-1-induced the capacity for vasculogenesis in vitro and in vivo. Moreover, CXCR7 gain of function markedly promoted vasculogenesis by MSCs in vitro and in vivo and induced endothelial differentiation along the arterial endothelial cell lineage via upregulation of Notch signaling. However, blockade of Notch signaling inhibited CXCR7-induced vasculogensis by MSCs. These results indicate CXCR7 is a critical regulator of MSC-mediated postnatal vasculogenesis and arterial specification via Notch signaling.
Collapse
|
21
|
Goodwin J, Laslett AL, Rugg-Gunn PJ. The application of cell surface markers to demarcate distinct human pluripotent states. Exp Cell Res 2020; 387:111749. [PMID: 31790696 PMCID: PMC6983944 DOI: 10.1016/j.yexcr.2019.111749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/17/2019] [Accepted: 11/27/2019] [Indexed: 01/24/2023]
Abstract
Recent advances in human pluripotent stem cell (hPSC) research have uncovered different subpopulations within stem cell cultures and have captured a range of pluripotent states that hold distinct molecular and functional properties. At the two ends of the pluripotency spectrum are naïve and primed hPSC, whereby naïve hPSC grown in stringent conditions recapitulate features of the preimplantation human embryo, and the conventionally grown primed hPSC align closer to the early postimplantation embryo. Investigating these cell types will help to define the mechanisms that control early development and should provide new insights into stem cell properties such as cell identity, differentiation and reprogramming. Monitoring cell surface marker expression provides a valuable approach to resolve complex cell populations, to directly compare between cell types, and to isolate viable cells for functional experiments. This review discusses the discovery and applications of cell surface markers to study human pluripotent cell types with a particular focus on the transitions between naïve and primed states. Highlighted areas for future study include the potential functions for the identified cell surface proteins in pluripotency, the production of new high-quality monoclonal antibodies to naïve-specific protein epitopes and the use of cell surface markers to characterise subpopulations within pluripotent states.
Collapse
Affiliation(s)
- Jacob Goodwin
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia; Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC 3800, Australia.
| | - Andrew L Laslett
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia; Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC 3800, Australia.
| | - Peter J Rugg-Gunn
- Epigenetics Programme, The Babraham Institute, Cambridge, UK; Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
22
|
Yu X, Liu H, Hamel KA, Morvan MG, Yu S, Leff J, Guan Z, Braz JM, Basbaum AI. Dorsal root ganglion macrophages contribute to both the initiation and persistence of neuropathic pain. Nat Commun 2020; 11:264. [PMID: 31937758 PMCID: PMC6959328 DOI: 10.1038/s41467-019-13839-2] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Paralleling the activation of dorsal horn microglia after peripheral nerve injury is a significant expansion and proliferation of macrophages around injured sensory neurons in dorsal root ganglia (DRG). Here we demonstrate a critical contribution of DRG macrophages, but not those at the nerve injury site, to both the initiation and maintenance of the mechanical hypersensitivity that characterizes the neuropathic pain phenotype. In contrast to the reported sexual dimorphism in the microglial contribution to neuropathic pain, depletion of DRG macrophages reduces nerve injury-induced mechanical hypersensitivity and expansion of DRG macrophages in both male and female mice. However, fewer macrophages are induced in the female mice and deletion of colony-stimulating factor 1 from sensory neurons, which prevents nerve injury-induced microglial activation and proliferation, only reduces macrophage expansion in male mice. Finally, we demonstrate molecular cross-talk between axotomized sensory neurons and macrophages, revealing potential peripheral DRG targets for neuropathic pain management. Interactions among spinal dorsal horn neurons and microglia contribute to the induction and maintenance of neuropathic pain after peripheral nerve injury. The authors show that depletion of macrophages in the dorsal root ganglia prevents and reverses ongoing nerve injury-induced hypersensitivity.
Collapse
Affiliation(s)
- Xiaobing Yu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA.
| | - Hongju Liu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA.,Department of Anesthesiology, Peking Union Medical College Hospital, Beijing, China
| | - Katherine A Hamel
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Maelig G Morvan
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Stephen Yu
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Jacqueline Leff
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, USA
| | - Joao M Braz
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Allan I Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
23
|
de Souza Lima IM, Schiavinato JLDS, Paulino Leite SB, Sastre D, Bezerra HLDO, Sangiorgi B, Corveloni AC, Thomé CH, Faça VM, Covas DT, Zago MA, Giacca M, Mano M, Panepucci RA. High-content screen in human pluripotent cells identifies miRNA-regulated pathways controlling pluripotency and differentiation. Stem Cell Res Ther 2019; 10:202. [PMID: 31287022 PMCID: PMC6615276 DOI: 10.1186/s13287-019-1318-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 06/11/2019] [Accepted: 06/30/2019] [Indexed: 01/13/2023] Open
Abstract
Background By post-transcriptionally regulating multiple target transcripts, microRNAs (miRNAs or miR) play important biological functions. H1 embryonic stem cells (hESCs) and NTera-2 embryonal carcinoma cells (ECCs) are two of the most widely used human pluripotent model cell lines, sharing several characteristics, including the expression of miRNAs associated to the pluripotent state or with differentiation. However, how each of these miRNAs functionally impacts the biological properties of these cells has not been systematically evaluated. Methods We investigated the effects of 31 miRNAs on NTera-2 and H1 hESCs, by transfecting miRNA mimics. Following 3–4 days of culture, cells were stained for the pluripotency marker OCT4 and the G2 cell-cycle marker Cyclin B1, and nuclei and cytoplasm were co-stained with Hoechst and Cell Mask Blue, respectively. By using automated quantitative fluorescence microscopy (i.e., high-content screening (HCS)), we obtained several morphological and marker intensity measurements, in both cell compartments, allowing the generation of a multiparametric miR-induced phenotypic profile describing changes related to proliferation, cell cycle, pluripotency, and differentiation. Results Despite the overall similarities between both cell types, some miRNAs elicited cell-specific effects, while some related miRNAs induced contrasting effects in the same cell. By identifying transcripts predicted to be commonly targeted by miRNAs inducing similar effects (profiles grouped by hierarchical clustering), we were able to uncover potentially modulated signaling pathways and biological processes, likely mediating the effects of the microRNAs on the distinct groups identified. Specifically, we show that miR-363 contributes to pluripotency maintenance, at least in part, by targeting NOTCH1 and PSEN1 and inhibiting Notch-induced differentiation, a mechanism that could be implicated in naïve and primed pluripotent states. Conclusions We present the first multiparametric high-content microRNA functional screening in human pluripotent cells. Integration of this type of data with similar data obtained from siRNA screenings (using the same HCS assay) could provide a large-scale functional approach to identify and validate microRNA-mediated regulatory mechanisms controlling pluripotency and differentiation. Electronic supplementary material The online version of this article (10.1186/s13287-019-1318-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ildercílio Mota de Souza Lima
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Josiane Lilian Dos Santos Schiavinato
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Sarah Blima Paulino Leite
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Danuta Sastre
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil
| | - Hudson Lenormando de Oliveira Bezerra
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Bruno Sangiorgi
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Amanda Cristina Corveloni
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Carolina Hassibe Thomé
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Marco Antônio Zago
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic and Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Miguel Mano
- Molecular Medicine Laboratory, International Centre for Genetic and Engineering and Biotechnology (ICGEB), Trieste, Italy.,Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Rodrigo Alexandre Panepucci
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil. .,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
24
|
Ashtari K, Nazari H, Ko H, Tebon P, Akhshik M, Akbari M, Alhosseini SN, Mozafari M, Mehravi B, Soleimani M, Ardehali R, Ebrahimi Warkiani M, Ahadian S, Khademhosseini A. Electrically conductive nanomaterials for cardiac tissue engineering. Adv Drug Deliv Rev 2019; 144:162-179. [PMID: 31176755 PMCID: PMC6784829 DOI: 10.1016/j.addr.2019.06.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 01/26/2023]
Abstract
Patient deaths resulting from cardiovascular diseases are increasing across the globe, posing the greatest risk to patients in developed countries. Myocardial infarction, as a result of inadequate blood flow to the myocardium, results in irreversible loss of cardiomyocytes which can lead to heart failure. A sequela of myocardial infarction is scar formation that can alter the normal myocardial architecture and result in arrhythmias. Over the past decade, a myriad of tissue engineering approaches has been developed to fabricate engineered scaffolds for repairing cardiac tissue. This paper highlights the recent application of electrically conductive nanomaterials (carbon and gold-based nanomaterials, and electroactive polymers) to the development of scaffolds for cardiac tissue engineering. Moreover, this work summarizes the effects of these nanomaterials on cardiac cell behavior such as proliferation and migration, as well as cardiomyogenic differentiation in stem cells.
Collapse
Affiliation(s)
- Khadijeh Ashtari
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran; Faculty of Advanced Technologies in Medicine, Department of Medical Nanotechnology, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hojjatollah Nazari
- Faculty of Advanced Technologies in Medicine, Department of Medical Nanotechnology, Iran University of Medical Sciences, Tehran, Iran; Stem Cell Technology Research Center, Tehran, Iran
| | - Hyojin Ko
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA
| | - Peyton Tebon
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA
| | - Masoud Akhshik
- Faculty of Forestry, University of Toronto, Toronto, Canada; Center for Biocomposites and Biomaterials Processing (CBBP), University of Toronto, Toronto, Canada; Shahdad Ronak Commercialization Company, Tehran, Iran
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, Canada; Center for Biomedical Research, University of Victoria, Victoria, Canada; Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, Canada
| | - Sanaz Naghavi Alhosseini
- Biomaterials Group, Department of Biomaterial Engineering, Amirkabir University of Technology, Tehran, Iran; Stem Cell Technology Research Center, Tehran, Iran
| | - Masoud Mozafari
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Bita Mehravi
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran; Faculty of Advanced Technologies in Medicine, Department of Medical Nanotechnology, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Faculty of Medical Sciences, Department of Hematology and Cell Therapy, Tarbiat Modares University, Tehran, Iran
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California - Los Angeles, USA
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia; Institute of Molecular Medicine, Sechenov University, Moscow, Russia
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, USA; Department of Bioengineering, University of California - Los Angeles, Los Angeles, USA; Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, USA; Department of Radiology, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, USA.
| |
Collapse
|
25
|
The Role of RNA Polymerase II Contiguity and Long-Range Interactions in the Regulation of Gene Expression in Human Pluripotent Stem Cells. Stem Cells Int 2019; 2019:1375807. [PMID: 30863449 PMCID: PMC6378007 DOI: 10.1155/2019/1375807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/05/2018] [Accepted: 12/24/2018] [Indexed: 12/02/2022] Open
Abstract
The eukaryotic nucleus is a highly complex structure that carries out multiple functions primarily needed for gene expression, and among them, transcription seems to be the most fundamental. Diverse approaches have demonstrated that transcription takes place at discrete sites known as transcription factories, wherein RNA polymerase II (RNAP II) is attached to the factory and immobilized while transcribing DNA. It has been proposed that transcription factories promote chromatin loop formation, creating long-range interactions in which relatively distant genes can be transcribed simultaneously. In this study, we examined long-range interactions between the POU5F1 gene and genes previously identified as being POU5F1 enhancer-interacting, namely, CDYL, TLE2, RARG, and MSX1 (all involved in transcriptional regulation), in human pluripotent stem cells (hPSCs) and their early differentiated counterparts. As a control gene, RUNX1 was used, which is expressed during hematopoietic differentiation and not associated with pluripotency. To reveal how these long-range interactions between POU5F1 and the selected genes change with the onset of differentiation and upon RNAP II inhibition, we performed three-dimensional fluorescence in situ hybridization (3D-FISH) followed by computational simulation analysis. Our analysis showed that the numbers of long-range interactions between specific genes decrease during differentiation, suggesting that the transcription of monitored genes is associated with pluripotency. In addition, we showed that upon inhibition of RNAP II, long-range associations do not disintegrate and remain constant. We also analyzed the distance distributions of these genes in the context of their positions in the nucleus and revealed that they tend to have similar patterns resembling normal distribution. Furthermore, we compared data created in vitro and in silico to assess the biological relevance of our results.
Collapse
|
26
|
MIB1 mutations reduce Notch signaling activation and contribute to congenital heart disease. Clin Sci (Lond) 2018; 132:2483-2491. [PMID: 30322850 PMCID: PMC6365626 DOI: 10.1042/cs20180732] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/07/2018] [Accepted: 10/11/2018] [Indexed: 01/12/2023]
Abstract
Congenital heart disease (CHD) is one of the most common birth defects in humans, but its genetic etiology remains largely unknown despite decades of research. The Notch signaling pathway plays critical roles in embryonic cardiogenesis. Mind bomb 1 (Mib1) is a vital protein that activates the Notch signaling pathway through promoting ubiquitination, endocytosis and subsequent activation of Notch ligands. Previous studies show that Mib1 knockout in mice completely abolishes Notch signaling, leading to cardiac deformity. However, the function of MIB1 and its potential disease-causing mutations are poorly studied in human CHD. In this research, we identified four novel non-synonymous heterozygous rare mutations of MIB1 from 417 Han Chinese CHD patients. The following biochemical analyses revealed that mutations p.T312K fs*55 and p.W271G significantly deplete MIB1’s function, resulting in a lower level of JAGGED1 (JAG1) ubiquitination and Notch signaling induction. Our results suggest that pathologic variants in MIB1 may contribute to CHD occurrence, shedding new light on the genetic mechanism of CHD in the context of the Notch signaling pathway.
Collapse
|
27
|
Fairhall EA, Leitch AC, Lakey AF, Probert PME, Richardson G, De Santis C, Wright MC. Glucocorticoid-induced pancreatic-hepatic trans-differentiation in a human cell line in vitro. Differentiation 2018; 102:10-18. [PMID: 29857331 DOI: 10.1016/j.diff.2018.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/02/2018] [Accepted: 05/21/2018] [Indexed: 11/28/2022]
Abstract
The rodent pancreatic AR42J-B13 (B-13) cell line differentiates into non-replicative hepatocyte-like cells in response to glucocorticoid mediated via the glucocorticoid receptor (GR). The aims of this study were to identify a human cell line that responds similarly and investigate the mechanisms underpinning any alteration in differentiation. Exposing the human pancreatic adenocarcinoma (HPAC) cell line to 1-10 µM concentrations of dexamethasone (DEX) resulted an inhibition of proliferation, suppressed carcinoembryonic antigen expression, limited expression of pancreatic acinar and hepatic gene expression and significant induction of the constitutively-expressed hepatic CYP3A5 mRNA transcript. These changes were associated with a pulse of genomic DNA methylation and suppressed notch signalling activity. HPAC cells expressed high levels of GR transcript in contrast to other nuclear receptors - such as the glucocorticoid-activated pregnane X receptor (PXR) - and GR transcriptional function was activated by DEX in HPAC cells. Expression of selected hepatocyte transcripts in response to DEX was blocked by co-treatment with the GR antagonist RU486. These data indicate that the HPAC response to glucocorticoid exposure includes an inhibition in proliferation, alterations in notch signalling and a limited change in the expression of genes associated with an acinar and hepatic phenotype. This is the first demonstration of a human cell responding to similarly to the rodent B-13 cell regarding formation of hepatocyte-like cells in response to glucocorticoid. Identifying and modulating the ablating factor(s) may enhance the hepatocyte-like forming capacity of HPAC cells after exposure to glucocorticoid and generate an unlimited in vitro supply of human hepatocytes for toxicology studies and a variety of clinical applications.
Collapse
Affiliation(s)
- Emma A Fairhall
- Institute of Cellular Medicine, Newcastle University, Level 4 William Leech Building, Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | - Alistair C Leitch
- Institute of Cellular Medicine, Newcastle University, Level 4 William Leech Building, Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | - Anne F Lakey
- Institute of Cellular Medicine, Newcastle University, Level 4 William Leech Building, Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | - Philip M E Probert
- Institute of Cellular Medicine, Newcastle University, Level 4 William Leech Building, Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | - Gabriella Richardson
- Institute of Cellular Medicine, Newcastle University, Level 4 William Leech Building, Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | - Carol De Santis
- Institute of Cellular Medicine, Newcastle University, Level 4 William Leech Building, Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| | - Matthew C Wright
- Institute of Cellular Medicine, Newcastle University, Level 4 William Leech Building, Medical School, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK.
| |
Collapse
|
28
|
Favarolo MB, López SL. Notch signaling in the division of germ layers in bilaterian embryos. Mech Dev 2018; 154:122-144. [PMID: 29940277 DOI: 10.1016/j.mod.2018.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 01/09/2023]
Abstract
Bilaterian embryos are triploblastic organisms which develop three complete germ layers (ectoderm, mesoderm, and endoderm). While the ectoderm develops mainly from the animal hemisphere, there is diversity in the location from where the endoderm and the mesoderm arise in relation to the animal-vegetal axis, ranging from endoderm being specified between the ectoderm and mesoderm in echinoderms, and the mesoderm being specified between the ectoderm and the endoderm in vertebrates. A common feature is that part of the mesoderm segregates from an ancient bipotential endomesodermal domain. The process of segregation is noisy during the initial steps but it is gradually refined. In this review, we discuss the role of the Notch pathway in the establishment and refinement of boundaries between germ layers in bilaterians, with special focus on its interaction with the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- María Belén Favarolo
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina
| | - Silvia L López
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina.
| |
Collapse
|
29
|
Li Y, Jin C, Bai H, Gao Y, Sun S, Chen L, Qin L, Liu PP, Cheng L, Wang QF. Human NOTCH4 is a key target of RUNX1 in megakaryocytic differentiation. Blood 2018; 131:191-201. [PMID: 29101237 PMCID: PMC5757696 DOI: 10.1182/blood-2017-04-780379] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022] Open
Abstract
Megakaryocytes (MKs) in adult marrow produce platelets that play important roles in blood coagulation and hemostasis. Monoallelic mutations of the master transcription factor gene RUNX1 lead to familial platelet disorder (FPD) characterized by defective MK and platelet development. However, the molecular mechanisms of FPD remain unclear. Previously, we generated human induced pluripotent stem cells (iPSCs) from patients with FPD containing a RUNX1 nonsense mutation. Production of MKs from the FPD-iPSCs was reduced, and targeted correction of the RUNX1 mutation restored MK production. In this study, we used isogenic pairs of FPD-iPSCs and the MK differentiation system to identify RUNX1 target genes. Using integrative genomic analysis of hematopoietic progenitor cells generated from FPD-iPSCs, and mutation-corrected isogenic controls, we identified 2 gene sets the transcription of which is either up- or downregulated by RUNX1 in mutation-corrected iPSCs. Notably, NOTCH4 expression was negatively controlled by RUNX1 via a novel regulatory DNA element within the locus, and we examined its involvement in MK generation. Specific inactivation of NOTCH4 by an improved CRISPR-Cas9 system in human iPSCs enhanced megakaryopoiesis. Moreover, small molecules known to inhibit Notch signaling promoted MK generation from both normal human iPSCs and postnatal CD34+ hematopoietic stem and progenitor cells. Our study newly identified NOTCH4 as a RUNX1 target gene and revealed a previously unappreciated role of NOTCH4 signaling in promoting human megakaryopoiesis. Our work suggests that human iPSCs with monogenic mutations have the potential to serve as an invaluable resource for discovery of novel druggable targets.
Collapse
Affiliation(s)
- Yueying Li
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Chen Jin
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Bai
- Division of Hematology, Department of Medicine and
- Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Yongxing Gao
- Division of Hematology, Department of Medicine and
- Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Shu Sun
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lei Chen
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lei Qin
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Paul P Liu
- Translational and Functional Genomics Branch, National Institutes of Health, National Human Genome Research Institute, Bethesda, MD
| | - Linzhao Cheng
- Division of Hematology, Department of Medicine and
- Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Qian-Fei Wang
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
Bigas A, Porcheri C. Notch and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:235-263. [DOI: 10.1007/978-3-319-89512-3_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Jung JH, Kang KW, Kim J, Hong SC, Park Y, Kim BS. CXCR2 Inhibition in Human Pluripotent Stem Cells Induces Predominant Differentiation to Mesoderm and Endoderm Through Repression of mTOR, β-Catenin, and hTERT Activities. Stem Cells Dev 2017; 25:1006-19. [PMID: 27188501 PMCID: PMC4931345 DOI: 10.1089/scd.2015.0395] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
On the basis of our previous report verifying that chemokine (C-X-C motif) receptor 2 (CXCR2) ligands in human placenta-derived cell conditioned medium (hPCCM) support human pluripotent stem cell (hPSC) propagation without exogenous basic fibroblast growth factor (bFGF), this study was designed to identify the effect of CXCR2 manipulation on the fate of hPSCs and the underlying mechanism, which had not been previously determined. We observed that CXCR2 inhibition in hPSCs induces predominant differentiation to mesoderm and endoderm with concomitant loss of hPSC characteristics and accompanying decreased expression of mammalian target of rapamycin (mTOR), β-catenin, and human telomerase reverse transcriptase (hTERT). These phenomena are recapitulated in hPSCs propagated in conventional culture conditions, including bFGF as well as those in hPCCM without exogenous bFGF, suggesting that the action of CXCR2 on hPSCs might not be associated with a bFGF-related mechanism. In addition, the specific CXCR2 ligand growth-related oncogene α (GROα) markedly increased the expression of ectodermal markers in differentiation-committed embryoid bodies derived from hPSCs. This finding suggests that CXCR2 inhibition in hPSCs prohibits the propagation of hPSCs and leads to predominant differentiation to mesoderm and endoderm owing to the blockage of ectodermal differentiation. Taken together, our results indicate that CXCR2 preferentially supports the maintenance of hPSC characteristics as well as facilitates ectodermal differentiation after the commitment to differentiation, and the mechanism might be associated with mTOR, β-catenin, and hTERT activities.
Collapse
Affiliation(s)
- Ji-Hye Jung
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,2 Department of Biomedical Science, Graduate School of Medicine, Korea University , Seoul, Korea
| | - Ka-Won Kang
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,3 Department of Hematology/Oncology, Korea University Anam Medical Center , Seoul, Korea
| | - Jihea Kim
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea
| | - Soon-Chul Hong
- 4 Department of Obstetrics/Gynecology, Korea University Anam Medical Center , Seoul, Korea
| | - Yong Park
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,3 Department of Hematology/Oncology, Korea University Anam Medical Center , Seoul, Korea
| | - Byung Soo Kim
- 1 Institute of Stem Cell Research, Korea University , Seoul, Korea.,2 Department of Biomedical Science, Graduate School of Medicine, Korea University , Seoul, Korea.,3 Department of Hematology/Oncology, Korea University Anam Medical Center , Seoul, Korea
| |
Collapse
|
32
|
C-terminal deletion of NOTCH1 intracellular domain (N1 ICD) increases its stability but does not amplify and recapitulate N1 ICD-dependent signalling. Sci Rep 2017; 7:5034. [PMID: 28698562 PMCID: PMC5506007 DOI: 10.1038/s41598-017-05119-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/24/2017] [Indexed: 12/16/2022] Open
Abstract
Since the generation of a mouse strain conditionally expressing the active intracellular domain of Notch1 (N1ICD), many laboratories have exploited this model (RosaN1-ICD) to assess the impact of constitutive Notch1 signalling activation in normal and pathological processes. It should be underscored that Cre-recombination leads to the expression of a C-terminally truncated form of N1ICD (N1ICDdC) in the RosaN1-ICD mutant mice. Given that no studies were undertaken to delineate whether deletion of this region leaves intact N1ICD function, stable cell lines with single targeted integration of inducible N1ICD and N1ICDdC were generated. We found that C-terminal deletion of N1ICD stabilized the protein but did not promote the activity of Notch responsive promoters. Furthermore, despite higher expression levels, N1ICDdC failed to phenocopy N1ICD in the promotion of anchorage-independent growth. Our results thus suggest that the C-terminal region of N1ICD plays a role in shaping the Notch response. Therefore, it should be taken into consideration that N1ICD is truncated when interpreting phenotypes of RosaN1-ICD mutant mice.
Collapse
|
33
|
Abstract
Cancer stem cells (CSCs), with their self-renewal ability and multilineage differentiation potential, are a critical subpopulation of tumor cells that can drive tumor initiation, growth, and resistance to therapy. Like embryonic and adult stem cells, CSCs express markers that are not expressed in normal somatic cells and are thus thought to contribute towards a 'stemness' phenotype. This review summarizes the current knowledge of stemness-related markers in human cancers, with a particular focus on important transcription factors, protein surface markers and signaling pathways.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Yvonne Li
- Dana Farber cancer Institute and Harvard Medical School, Boston, Massachusetts 02115
| | - Xun Zhang
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
34
|
Hrstka SCL, Li X, Nelson TJ. NOTCH1-Dependent Nitric Oxide Signaling Deficiency in Hypoplastic Left Heart Syndrome Revealed Through Patient-Specific Phenotypes Detected in Bioengineered Cardiogenesis. Stem Cells 2017; 35:1106-1119. [PMID: 28142228 DOI: 10.1002/stem.2582] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 11/12/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022]
Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart defect (CHD) attributable to multifactorial molecular underpinnings. Multiple genetic loci have been implicated to increase the risk of disease, yet genotype-phenotype relationships remain poorly defined. Whole genome sequencing complemented by cardiac phenotype from five individuals in an HLHS-affected family enabled the identification of NOTCH1 as a prioritized candidate gene linked to CHD in three individuals with mutant allele burden significantly impairing Notch signaling in the HLHS-affected proband. To better understand a mechanistic basis through which NOTCH1 contributes to heart development, human induced pluripotent stem cells (hiPSCs) were created from the HLHS-affected parent-proband triad and differentiated into cardiovascular cell lineages for molecular characterization. HLHS-affected hiPSCs exhibited a deficiency in Notch signaling pathway components and a diminished capacity to generate hiPSC-cardiomyocytes. Optimization of conditions to procure HLHS-hiPSC-cardiomyocytes led to an approach that compensated for dysregulated nitric oxide (NO)-dependent Notch signaling in the earliest specification stages. Augmentation of HLHS-hiPSCs with small molecules stimulating NO signaling in the first 4 days of differentiation provided a cardiomyocyte yield equivalent to the parental hiPSCs. No discernable differences in calcium dynamics were observed between the bioengineered cardiomyocytes derived from the proband and the parents. We conclude that in vitro modeling with HLHS-hiPSCs bearing NOTCH1 mutations facilitated the discovery of a NO-dependent signaling component essential for cardiovascular cell lineage specification. Potentiation of NO signaling with small therapeutic molecules restored cardiogenesis in vitro and may identify a potential therapeutic target for patients affected by functionally compromised NOTCH1 variants. Stem Cells 2017;35:1106-1119.
Collapse
Affiliation(s)
- Sybil C L Hrstka
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Xing Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA.,Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy J Nelson
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,General Internal Medicine and Transplant Center, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
35
|
Gamage TK, Chamley LW, James JL. Stem cell insights into human trophoblast lineage differentiation. Hum Reprod Update 2016; 23:77-103. [PMID: 27591247 DOI: 10.1093/humupd/dmw026] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/27/2016] [Accepted: 07/05/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The human placenta is vital for fetal development, yet little is understood about how it forms successfully to ensure a healthy pregnancy or why this process is inadequate in 1 in 10 pregnancies, leading to miscarriage, intrauterine growth restriction or preeclampsia. Trophoblasts are placenta-specific epithelial cells that maximize nutrient exchange. All trophoblast lineages are thought to arise from a population of trophoblast stem cells (TSCs). However, whilst the isolation of murine TSC has led to an explosion in understanding murine placentation, the isolation of an analogous human TSC has proved more difficult. Consequently, alternative methods of studying human trophoblast lineage development have been employed, including human embryonic stem cells (hESCs), induced pluripotent stem cells (iPS) and transformed cell lines; but what do these proxy models tell us about what is happening during early placental development? OBJECTIVE AND RATIONALE In this systematic review, we evaluate current approaches to understanding human trophoblast lineage development in order to collate and refine these models and inform future approaches aimed at establishing human TSC lines. SEARCH METHODS To ensure all relevant articles were analysed, an unfiltered search of Pubmed, Embase, Scopus and Web of Science was conducted for 25 key terms on the 13th May 2016. In total, 47 313 articles were retrieved and manually filtered based on non-human, non-English, non-full text, non-original article and off-topic subject matter. This resulted in a total of 71 articles deemed relevant for review in this article. OUTCOMES Candidate human TSC populations have been identified in, and isolated from, both the chorionic membrane and villous tissue of the placenta, but further investigation is required to validate these as 'true' human TSCs. Isolating human TSCs from blastocyst trophectoderm has not been successful in humans as it was in mice, although recently the first reported TSC line (USFB6) was isolated from an eight-cell morula. In lieu of human TSC lines, trophoblast-like cells have been induced to differentiate from hESCs and iPS. However, differentiation in these model systems is difficult to control, culture conditions employed are highly variable, and the extent to which they accurately convey the biology of 'true' human TSCs remains unclear, particularly as a consensus has not been met among the scientific community regarding which characteristics a human TSC must possess. WIDER IMPLICATIONS Human TSC models have the potential to revolutionize our understanding of trophoblast differentiation, allowing us to make significant gains in understanding the underlying pathology of pregnancy disorders and to test potential therapeutic interventions on cell function in vitro. In order to do this, a collaborative effort is required to establish the criteria that define a human TSC to confirm the presence of human TSCs in both primary isolates and to determine how accurately trophoblast-like cells derived from current model systems reflect trophoblast from primary tissue. The in vitro systems currently used to model early trophoblast lineage formation have provided insights into early human placental formation but it is unclear whether these trophoblast-like cells are truly representative of primary human trophoblast. Consequently, continued refinement of current models, and standardization of culture protocols is essential to aid our ability to identify, isolate and propagate 'true' human TSCs from primary tissue.
Collapse
Affiliation(s)
- Teena Kjb Gamage
- Department of Obstetrics and Gynaecology, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynaecology, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
36
|
Majumder M, Xin X, Liu L, Tutunea-Fatan E, Rodriguez-Torres M, Vincent K, Postovit LM, Hess D, Lala PK. COX-2 Induces Breast Cancer Stem Cells via EP4/PI3K/AKT/NOTCH/WNT Axis. Stem Cells 2016; 34:2290-2305. [PMID: 27301070 DOI: 10.1002/stem.2426] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 04/04/2016] [Accepted: 04/18/2016] [Indexed: 01/03/2025]
Abstract
Cancer stem-like cells (SLC) resist conventional therapies, necessitating searches for SLC-specific targets. We established that cyclo-oxygenase(COX)-2 expression promotes human breast cancer progression by activation of the prostaglandin(PG)E-2 receptor EP4. Present study revealed that COX-2 induces SLCs by EP4-mediated NOTCH/WNT signaling. Ectopic COX-2 over-expression in MCF-7 and SKBR-3 cell lines resulted in: increased migration/invasion/proliferation, epithelial-mesenchymal transition (EMT), elevated SLCs (spheroid formation), increased ALDH activity and colocalization of COX-2 and SLC markers (ALDH1A, CD44, β-Catenin, NANOG, OCT3/4, SOX-2) in spheroids. These changes were reversed with COX-2-inhibitor or EP4-antagonist (EP4A), indicating dependence on COX-2/EP4 activities. COX-2 over-expression or EP4-agonist treatments of COX-2-low cells caused up-regulation of NOTCH/WNT genes, blocked with PI3K/AKT inhibitors. NOTCH/WNT inhibitors also blocked COX-2/EP4 induced SLC induction. Microarray analysis showed up-regulation of numerous SLC-regulatory and EMT-associated genes. MCF-7-COX-2 cells showed increased mammary tumorigenicity and spontaneous multiorgan metastases in NOD/SCID/IL-2Rγ-null mice for successive generations with limiting cell inocula. These tumors showed up-regulation of VEGF-A/C/D, Vimentin and phospho-AKT, down-regulation of E-Cadherin and enrichment of SLC marker positive and spheroid forming cells. MCF-7-COX-2 cells also showed increased lung colonization in NOD/SCID/GUSB-null mice, an effect reversed with EP4-knockdown or EP4A treatment of the MCF-7-COX-2 cells. COX-2/EP4/ALDH1A mRNA expression in human breast cancer tissues were highly correlated with one other, more marked in progressive stage of disease. In situ immunostaining of human breast tumor tissues revealed co-localization of SLC markers with COX-2, supporting COX-2 inducing SLCs. High COX-2/EP4 mRNA expression was linked with reduced survival. Thus, EP4 represents a novel SLC-ablative target in human breast cancer. Stem Cells 2016;34:2290-2305.
Collapse
Affiliation(s)
| | | | - Ling Liu
- Department of Anatomy and Cell Biology
| | | | | | - Krista Vincent
- Department of Anatomy and Cell Biology
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Lynne-Marie Postovit
- Department of Anatomy and Cell Biology
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - David Hess
- Physiology and Pharmacology
- Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada
| | - Peeyush K Lala
- Department of Anatomy and Cell Biology.
- Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
37
|
Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as a Model for Heart Development and Congenital Heart Disease. Stem Cell Rev Rep 2016; 11:710-27. [PMID: 26085192 DOI: 10.1007/s12015-015-9596-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Congenital heart disease (CHD) remains a significant health problem, with a growing population of survivors with chronic disease. Despite intense efforts to understand the genetic basis of CHD in humans, the etiology of most CHD is unknown. Furthermore, new models of CHD are required to better understand the development of CHD and to explore novel therapies for this patient population. In this review, we highlight the role that human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes can serve to enhance our understanding of the development, pathophysiology and potential therapeutic targets for CHD. We highlight the use of hiPSC-derived cardiomyocytes to model gene regulatory interactions, cell-cell interactions and tissue interactions contributing to CHD. We further emphasize the importance of using hiPSC-derived cardiomyocytes as personalized research models. The use of hiPSCs presents an unprecedented opportunity to generate disease-specific cellular models, investigate the underlying molecular mechanisms of disease and uncover new therapeutic targets for CHD.
Collapse
|
38
|
Welte T, Kim IS, Tian L, Gao X, Wang H, Li J, Holdman XB, Herschkowitz JI, Pond A, Xie G, Kurley S, Nguyen T, Liao L, Dobrolecki LE, Mo Q, Edwards DP, Huang S, Xin L, Xu J, Li Y, Lewis MT, Wang T, Westbrook TF, Rosen JM, Zhang XHF. Oncogenic mTOR signalling recruits myeloid-derived suppressor cells to promote tumour initiation. Nat Cell Biol 2016; 18:632-44. [PMID: 27183469 PMCID: PMC4884142 DOI: 10.1038/ncb3355] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/11/2016] [Indexed: 12/14/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) play critical roles in primary and metastatic cancer progression. MDSC regulation is widely variable even among patients harbouring the same type of malignancy, and the mechanisms governing such heterogeneity are largely unknown. Here, integrating human tumour genomics and syngeneic mammary tumour models, we demonstrate that mTOR signalling in cancer cells dictates a mammary tumour's ability to stimulate MDSC accumulation through regulating G-CSF. Inhibiting this pathway or its activators (for example, FGFR) impairs tumour progression, which is partially rescued by restoring MDSCs or G-CSF. Tumour-initiating cells (TICs) exhibit elevated G-CSF. MDSCs reciprocally increase TIC frequency through activating Notch in tumour cells, forming a feedforward loop. Analyses of primary breast cancers and patient-derived xenografts corroborate these mechanisms in patients. These findings establish a non-canonical oncogenic role of mTOR signalling in recruiting pro-tumorigenic MDSCs and show how defined cancer subsets may evolve to promote and depend on a distinct immune microenvironment.
Collapse
Affiliation(s)
- Thomas Welte
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Diana Helis Henry Medical Research Foundation, New Orlean, LA 70130, USA
| | - Ik Sun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Lin Tian
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Xia Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - June Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Xue B. Holdman
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Jason I. Herschkowitz
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Adam Pond
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | | | - Sarah Kurley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Tuan Nguyen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Qianxing Mo
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Dean P. Edwards
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Shixia Huang
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Li Xin
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Jianming Xu
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Tian Wang
- Department of Microbiology & Immunology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555
| | - Thomas F. Westbrook
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Jeffrey M. Rosen
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
- McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| |
Collapse
|
39
|
A dual molecular analogue tuner for dissecting protein function in mammalian cells. Nat Commun 2016; 7:11742. [PMID: 27230261 PMCID: PMC4895048 DOI: 10.1038/ncomms11742] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/26/2016] [Indexed: 12/16/2022] Open
Abstract
Loss-of-function studies are fundamental for dissecting gene function. Yet, methods
to rapidly and effectively perturb genes in mammalian cells, and particularly in
stem cells, are scarce. Here we present a system for simultaneous conditional
regulation of two different proteins in the same mammalian cell. This system
harnesses the plant auxin and jasmonate hormone-induced degradation pathways, and is
deliverable with only two lentiviral vectors. It combines RNAi-mediated silencing of
two endogenous proteins with the expression of two exogenous proteins whose
degradation is induced by external ligands in a rapid, reversible, titratable and
independent manner. By engineering molecular tuners for NANOG, CHK1, p53 and NOTCH1
in mammalian stem cells, we have validated the applicability of the system and
demonstrated its potential to unravel complex biological processes. Loss-of-function approaches are fundamental for dissecting the roles
played by genes but methods to simultaneously perturb several proteins in the same
mammalian cell are scarce. Here the authors harness the plant auxin and jasmonate
hormone-degradation pathways and RNAi technology, to control the levels of two proteins
and validate its application in stem cells.
Collapse
|
40
|
Souilhol C, Perea-Gomez A, Camus A, Beck-Cormier S, Vandormael-Pournin S, Escande M, Collignon J, Cohen-Tannoudji M. NOTCH activation interferes with cell fate specification in the gastrulating mouse embryo. Development 2016; 142:3649-60. [PMID: 26534985 DOI: 10.1242/dev.121145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
NOTCH signalling is an evolutionarily conserved pathway involved in intercellular communication essential for cell fate choices during development. Although dispensable for early aspects of mouse development, canonical RBPJ-dependent NOTCH signalling has been shown to influence lineage commitment during embryonic stem cell (ESC) differentiation. NOTCH activation in ESCs promotes the acquisition of a neural fate, whereas its suppression favours their differentiation into cardiomyocytes. This suggests that NOTCH signalling is implicated in the acquisition of distinct embryonic fates at early stages of mammalian development. In order to investigate in vivo such a role for NOTCH signalling in shaping cell fate specification, we use genetic approaches to constitutively activate the NOTCH pathway in the mouse embryo. Early embryonic development, including the establishment of anterior-posterior polarity, is not perturbed by forced NOTCH activation. By contrast, widespread NOTCH activity in the epiblast triggers dramatic gastrulation defects. These are fully rescued in a RBPJ-deficient background. Epiblast-specific NOTCH activation induces acquisition of neurectoderm identity and disrupts the formation of specific mesodermal precursors including the derivatives of the anterior primitive streak, the mouse organiser. In addition, we show that forced NOTCH activation results in misregulation of NODAL signalling, a major determinant of early embryonic patterning. Our study reveals a previously unidentified role for canonical NOTCH signalling during mammalian gastrulation. It also exemplifies how in vivo studies can shed light on the mechanisms underlying cell fate specification during in vitro directed differentiation.
Collapse
Affiliation(s)
- Céline Souilhol
- Institut Pasteur, Unité de Génétique Fonctionnelle de la Souris, Département de Biologie du Développement et Cellules Souches, 25 rue du docteur Roux, Paris F-75015, France CNRS URA 2578, Paris F-75015, France
| | - Aitana Perea-Gomez
- Institut Jacques Monod, CNRS, UMR7592, Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Anne Camus
- Institut Jacques Monod, CNRS, UMR7592, Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Sarah Beck-Cormier
- Institut Pasteur, Unité de Génétique Fonctionnelle de la Souris, Département de Biologie du Développement et Cellules Souches, 25 rue du docteur Roux, Paris F-75015, France CNRS URA 2578, Paris F-75015, France
| | - Sandrine Vandormael-Pournin
- Institut Pasteur, Unité de Génétique Fonctionnelle de la Souris, Département de Biologie du Développement et Cellules Souches, 25 rue du docteur Roux, Paris F-75015, France CNRS URA 2578, Paris F-75015, France
| | - Marie Escande
- Institut Pasteur, Unité de Génétique Fonctionnelle de la Souris, Département de Biologie du Développement et Cellules Souches, 25 rue du docteur Roux, Paris F-75015, France CNRS URA 2578, Paris F-75015, France
| | - Jérôme Collignon
- Institut Jacques Monod, CNRS, UMR7592, Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75205, France
| | - Michel Cohen-Tannoudji
- Institut Pasteur, Unité de Génétique Fonctionnelle de la Souris, Département de Biologie du Développement et Cellules Souches, 25 rue du docteur Roux, Paris F-75015, France CNRS URA 2578, Paris F-75015, France
| |
Collapse
|
41
|
Hadjimichael C, Chanoumidou K, Papadopoulou N, Arampatzi P, Papamatheakis J, Kretsovali A. Common stemness regulators of embryonic and cancer stem cells. World J Stem Cells 2015; 7:1150-1184. [PMID: 26516408 PMCID: PMC4620423 DOI: 10.4252/wjsc.v7.i9.1150] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/30/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023] Open
Abstract
Pluripotency of embryonic stem cells (ESCs) and induced pluripotent stem cells is regulated by a well characterized gene transcription circuitry. The circuitry is assembled by ESC specific transcription factors, signal transducing molecules and epigenetic regulators. Growing understanding of stem-like cells, albeit of more complex phenotypes, present in tumors (cancer stem cells), provides a common conceptual and research framework for basic and applied stem cell biology. In this review, we highlight current results on biomarkers, gene signatures, signaling pathways and epigenetic regulators that are common in embryonic and cancer stem cells. We discuss their role in determining the cell phenotype and finally, their potential use to design next generation biological and pharmaceutical approaches for regenerative medicine and cancer therapies.
Collapse
|
42
|
Compound heterozygous NOTCH1 mutations underlie impaired cardiogenesis in a patient with hypoplastic left heart syndrome. Hum Genet 2015; 134:1003-11. [DOI: 10.1007/s00439-015-1582-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/02/2015] [Indexed: 12/24/2022]
|
43
|
Carvalho FLF, Marchionni L, Gupta A, Kummangal BA, Schaeffer EM, Ross AE, Berman DM. HES6 promotes prostate cancer aggressiveness independently of Notch signalling. J Cell Mol Med 2015; 19:1624-36. [PMID: 25864518 PMCID: PMC4511360 DOI: 10.1111/jcmm.12537] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 12/17/2014] [Indexed: 12/22/2022] Open
Abstract
Notch signalling is implicated in the pathogenesis of a variety of cancers, but its role in prostate cancer is poorly understood. However, selected Notch pathway members are overrepresented in high-grade prostate cancers. We comprehensively profiled Notch pathway components in prostate cells and found prostate cancer-specific up-regulation of NOTCH3 and HES6. Their expression was particularly high in androgen responsive lines. Up- and down-regulating Notch in these cells modulated expression of canonical Notch targets, HES1 and HEY1, which could also be induced by androgen. Surprisingly, androgen treatment also suppressed Notch receptor expression, suggesting that androgens can activate Notch target genes in a receptor-independent manner. Using a Notch-sensitive Recombination signal binding protein for immunoglobulin kappa J region (RBPJ) reporter assay, we found that basal levels of Notch signalling were significantly lower in prostate cancer cells compared to benign cells. Accordingly pharmacological Notch pathway blockade did not inhibit cancer cell growth or viability. In contrast to canonical Notch targets, HES6, a HES family member known to antagonize Notch signalling, was not regulated by Notch signalling, but relied instead on androgen levels, both in cultured cells and in human cancer tissues. When engineered into prostate cancer cells, reduced levels of HES6 resulted in reduced cancer cell invasion and clonogenic growth. By molecular profiling, we identified potential roles for HES6 in regulating hedgehog signalling, apoptosis and cell migration. Our results did not reveal any cell-autonomous roles for canonical Notch signalling in prostate cancer. However, the results do implicate HES6 as a promoter of prostate cancer progression.
Collapse
Affiliation(s)
- Filipe L F Carvalho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luigi Marchionni
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anuj Gupta
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Basheer A Kummangal
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward M Schaeffer
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Brady Institute of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ashley E Ross
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Brady Institute of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David M Berman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Brady Institute of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Pathology and Molecular Medicine and Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
| |
Collapse
|
44
|
Ventre S, Indrieri A, Fracassi C, Franco B, Conte I, Cardone L, di Bernardo D. Metabolic regulation of the ultradian oscillator Hes1 by reactive oxygen species. J Mol Biol 2015; 427:1887-902. [PMID: 25796437 DOI: 10.1016/j.jmb.2015.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/07/2015] [Accepted: 03/11/2015] [Indexed: 12/25/2022]
Abstract
Ultradian oscillators are cyclically expressed genes with a period of less than 24h, found in the major signalling pathways. The Notch effector hairy and enhancer of split Hes genes are ultradian oscillators. The physiological signals that synchronise and entrain Hes oscillators remain poorly understood. We investigated whether cellular metabolism modulates Hes1 cyclic expression. We demonstrated that, in mouse myoblasts (C2C12), Hes1 oscillation depends on reactive oxygen species (ROS), which are generated by the mitochondria electron transport chain and by NADPH oxidases NOXs. In vitro, the regulation of Hes1 by ROS occurs via the calcium-mediated signalling. The modulation of Hes1 by ROS was relevant in vivo, since perturbing ROS homeostasis was sufficient to alter Medaka (Oryzias latipes) somitogenesis, a process that is dependent on Hes1 ultradian oscillation during embryo development. Moreover, in a Medaka model for human microphthalmia with linear skin lesions syndrome, in which mitochondrial ROS homeostasis was impaired, we documented important somitogenesis defects and the deregulation of Hes homologues genes involved in somitogenesis. Notably, both molecular and developmental defects were rescued by antioxidant treatments. Our studies provide the first evidence of a coupling between cellular redox metabolism and an ultradian biological oscillator with important pathophysiological implication for somitogenesis.
Collapse
Affiliation(s)
- Simona Ventre
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078 Naples, Italy
| | - Alessia Indrieri
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078 Naples, Italy
| | - Chiara Fracassi
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078 Naples, Italy
| | - Brunella Franco
- Department of Medical Translational Sciences, University of Naples Federico II, 80138 Napoli, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078 Naples, Italy
| | - Luca Cardone
- Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy.
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078 Naples, Italy; Department of Electrical Engineering and Information Technology, University of Naples Federico II, 80138 Napoli, Italy.
| |
Collapse
|
45
|
Ayllón V, Bueno C, Ramos-Mejía V, Navarro-Montero O, Prieto C, Real PJ, Romero T, García-León MJ, Toribio ML, Bigas A, Menendez P. The Notch ligand DLL4 specifically marks human hematoendothelial progenitors and regulates their hematopoietic fate. Leukemia 2015; 29:1741-53. [PMID: 25778099 DOI: 10.1038/leu.2015.74] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 03/08/2015] [Accepted: 03/09/2015] [Indexed: 12/17/2022]
Abstract
Notch signaling is essential for definitive hematopoiesis, but its role in human embryonic hematopoiesis is largely unknown. We show that in hESCs the expression of the Notch ligand DLL4 is induced during hematopoietic differentiation. We found that DLL4 is only expressed in a sub-population of bipotent hematoendothelial progenitors (HEPs) and segregates their hematopoietic versus endothelial potential. We demonstrate at the clonal level and through transcriptome analyses that DLL4(high) HEPs are enriched in endothelial potential, whereas DLL4(low/-) HEPs are committed to the hematopoietic lineage, albeit both populations still contain bipotent cells. Moreover, DLL4 stimulation enhances hematopoietic differentiation of HEPs and increases the amount of clonogenic hematopoietic progenitors. Confocal microscopy analysis of whole differentiating embryoid bodies revealed that DLL4(high) HEPs are located close to DLL4(low/-) HEPs, and at the base of clusters of CD45+ cells, resembling intra-aortic hematopoietic clusters found in mouse embryos. We propose a model for human embryonic hematopoiesis in which DLL4(low/-) cells within hemogenic endothelium receive Notch-activating signals from DLL4(high) cells, resulting in an endothelial-to-hematopoietic transition and their differentiation into CD45+ hematopoietic cells.
Collapse
Affiliation(s)
- V Ayllón
- Gene Regulation, Stem Cells & Development Laboratory, GENyO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - C Bueno
- Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Barcelona, Spain
| | - V Ramos-Mejía
- Gene Regulation, Stem Cells & Development Laboratory, GENyO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - O Navarro-Montero
- Gene Regulation, Stem Cells & Development Laboratory, GENyO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - C Prieto
- Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Barcelona, Spain
| | - P J Real
- Gene Regulation, Stem Cells & Development Laboratory, GENyO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - T Romero
- Gene Regulation, Stem Cells & Development Laboratory, GENyO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - M J García-León
- Centro de Biologia Molecular Severo Ochoa (CBM-SO), CSIC-UAM, Campus de la Universidad Autonoma de Madrid, Madrid, Spain
| | - M L Toribio
- Centro de Biologia Molecular Severo Ochoa (CBM-SO), CSIC-UAM, Campus de la Universidad Autonoma de Madrid, Madrid, Spain
| | - A Bigas
- Program in Cancer Research, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - P Menendez
- 1] Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Barcelona, Spain [2] Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
46
|
D'Angelo RC, Ouzounova M, Davis A, Choi D, Tchuenkam SM, Kim G, Luther T, Quraishi AA, Senbabaoglu Y, Conley SJ, Clouthier SG, Hassan KA, Wicha MS, Korkaya H. Notch reporter activity in breast cancer cell lines identifies a subset of cells with stem cell activity. Mol Cancer Ther 2015; 14:779-787. [PMID: 25673823 DOI: 10.1158/1535-7163.mct-14-0228] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 12/08/2014] [Indexed: 12/12/2022]
Abstract
Developmental pathways such as Notch play a pivotal role in tissue-specific stem cell self-renewal as well as in tumor development. However, the role of Notch signaling in breast cancer stem cells (CSC) remains to be determined. We utilized a lentiviral Notch reporter system to identify a subset of cells with a higher Notch activity (Notch(+)) or reduced activity (Notch(-)) in multiple breast cancer cell lines. Using in vitro and mouse xenotransplantation assays, we investigated the role of the Notch pathway in breast CSC regulation. Breast cancer cells with increased Notch activity displayed increased sphere formation as well as expression of breast CSC markers. Interestingly Notch(+) cells displayed higher Notch4 expression in both basal and luminal breast cancer cell lines. Moreover, Notch(+) cells demonstrated tumor initiation capacity at serial dilutions in mouse xenografts, whereas Notch(-) cells failed to generate tumors. γ-Secretase inhibitor (GSI), a Notch blocker but not a chemotherapeutic agent, effectively targets these Notch(+) cells in vitro and in mouse xenografts. Furthermore, elevated Notch4 and Hey1 expression in primary patient samples correlated with poor patient survival. Our study revealed a molecular mechanism for the role of Notch-mediated regulation of breast CSCs and provided a compelling rationale for CSC-targeted therapeutics.
Collapse
Affiliation(s)
| | | | | | - Daejin Choi
- Georgia Regents University Cancer Center, Department of Biochemistry and Molecular Biology 1410 Laney Walker Blvd. CN2136 Augusta, GA 30912
| | - Stevie M Tchuenkam
- Georgia Regents University Cancer Center, Department of Biochemistry and Molecular Biology 1410 Laney Walker Blvd. CN2136 Augusta, GA 30912
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sarkar P, Randall SM, Collier TS, Nero A, Russell TA, Muddiman DC, Rao BM. Activin/nodal signaling switches the terminal fate of human embryonic stem cell-derived trophoblasts. J Biol Chem 2015; 290:8834-48. [PMID: 25670856 DOI: 10.1074/jbc.m114.620641] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Indexed: 11/06/2022] Open
Abstract
Human embryonic stem cells (hESCs) have been routinely treated with bone morphogenetic protein and/or inhibitors of activin/nodal signaling to obtain cells that express trophoblast markers. Trophoblasts can terminally differentiate to either extravillous trophoblasts or syncytiotrophoblasts. The signaling pathways that govern the terminal fate of these trophoblasts are not understood. We show that activin/nodal signaling switches the terminal fate of these hESC-derived trophoblasts. Inhibition of activin/nodal signaling leads to formation of extravillous trophoblast, whereas loss of activin/nodal inhibition leads to the formation of syncytiotrophoblasts. Also, the ability of hESCs to form bona fide trophoblasts has been intensely debated. We have examined hESC-derived trophoblasts in the light of stringent criteria that were proposed recently, such as hypomethylation of the ELF5-2b promoter region and down-regulation of HLA class I antigens. We report that trophoblasts that possess these properties can indeed be obtained from hESCs.
Collapse
Affiliation(s)
| | - Shan M Randall
- the W. M. Keck FT-ICR Mass Spectrometry Laboratory, Department of Chemistry, and
| | - Timothy S Collier
- the W. M. Keck FT-ICR Mass Spectrometry Laboratory, Department of Chemistry, and
| | - Anthony Nero
- From the Department of Chemical and Biomolecular Engineering
| | - Teal A Russell
- the Department of Biochemistry, North Carolina State University, Raleigh, North Carolina 27695
| | - David C Muddiman
- the W. M. Keck FT-ICR Mass Spectrometry Laboratory, Department of Chemistry, and
| | - Balaji M Rao
- From the Department of Chemical and Biomolecular Engineering,
| |
Collapse
|
48
|
Guiu J, Bergen DJM, De Pater E, Islam ABMMK, Ayllón V, Gama-Norton L, Ruiz-Herguido C, González J, López-Bigas N, Menendez P, Dzierzak E, Espinosa L, Bigas A. Identification of Cdca7 as a novel Notch transcriptional target involved in hematopoietic stem cell emergence. ACTA ACUST UNITED AC 2014; 211:2411-23. [PMID: 25385755 PMCID: PMC4235648 DOI: 10.1084/jem.20131857] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Guiu et al. use ChIP-on-chip analysis for the Notch partner RBPj, using embryonic tissue from the aorta-gonad-mesonephros region to identify potential novel Notch target genes involved in HSC emergence. They show that c-MYC–responsive gene Cdca7 is expressed in different HSC and progenitor subpopulations and that CDCA7 is important for maintaining the undifferentiated phenotype. Cdca7 acts downstream of Notch in HSCs in zebrafish, mouse, and human, indicating a highly conserved Notch/RBPj/Cdca7 axis in hematopoietic development. Hematopoietic stem cell (HSC) specification occurs in the embryonic aorta and requires Notch activation; however, most of the Notch-regulated elements controlling de novo HSC generation are still unknown. Here, we identify putative direct Notch targets in the aorta-gonad-mesonephros (AGM) embryonic tissue by chromatin precipitation using antibodies against the Notch partner RBPj. By ChIP-on-chip analysis of the precipitated DNA, we identified 701 promoter regions that were candidates to be regulated by Notch in the AGM. One of the most enriched regions corresponded to the Cdca7 gene, which was subsequently confirmed to recruit the RBPj factor but also Notch1 in AGM cells. We found that during embryonic hematopoietic development, expression of Cdca7 is restricted to the hematopoietic clusters of the aorta, and it is strongly up-regulated in the hemogenic population during human embryonic stem cell hematopoietic differentiation in a Notch-dependent manner. Down-regulation of Cdca7 mRNA in cultured AGM cells significantly induces hematopoietic differentiation and loss of the progenitor population. Finally, using loss-of-function experiments in zebrafish, we demonstrate that CDCA7 contributes to HSC emergence in vivo during embryonic development. Thus, our study identifies Cdca7 as an evolutionary conserved Notch target involved in HSC emergence.
Collapse
Affiliation(s)
- Jordi Guiu
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Dylan J M Bergen
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Emma De Pater
- Erasmus MC Stem Cell and Regenerative Medicine Institute, Erasmus Medical Center, 3000 CA Rotterdam, Netherlands
| | - Abul B M M K Islam
- Research Unit on Biomedical Informatics, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Verónica Ayllón
- Centre for Genomics and Oncological Research (Genyo), Pfizer-University of Granada-Andalusian Government, 18016 Granada, Spain
| | - Leonor Gama-Norton
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Cristina Ruiz-Herguido
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Jessica González
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Nuria López-Bigas
- Research Unit on Biomedical Informatics, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Pablo Menendez
- José Carreras Leukaemia Research Institute, Cell Therapy Program, School of Medicine, University of Barcelona, 08036 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Elaine Dzierzak
- Erasmus MC Stem Cell and Regenerative Medicine Institute, Erasmus Medical Center, 3000 CA Rotterdam, Netherlands
| | - Lluis Espinosa
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| | - Anna Bigas
- Program de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain
| |
Collapse
|
49
|
Kushwah R, Guezguez B, Lee JB, Hopkins CI, Bhatia M. Pleiotropic roles of Notch signaling in normal, malignant, and developmental hematopoiesis in the human. EMBO Rep 2014; 15:1128-38. [PMID: 25252682 DOI: 10.15252/embr.201438842] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Notch signaling pathway is evolutionarily conserved across species and plays an important role in regulating cell differentiation, proliferation, and survival. It has been implicated in several different hematopoietic processes including early hematopoietic development as well as adult hematological malignancies in humans. This review focuses on recent developments in understanding the role of Notch signaling in the human hematopoietic system with an emphasis on hematopoietic initiation from human pluripotent stem cells and regulation within the bone marrow. Based on recent insights, we summarize potential strategies for treatment of human hematological malignancies toward the concept of targeting Notch signaling for fate regulation.
Collapse
Affiliation(s)
- Rahul Kushwah
- McMaster Stem Cell and Cancer Research Institute (SCC-RI), Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Borhane Guezguez
- McMaster Stem Cell and Cancer Research Institute (SCC-RI), Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Jung Bok Lee
- McMaster Stem Cell and Cancer Research Institute (SCC-RI), Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Claudia I Hopkins
- McMaster Stem Cell and Cancer Research Institute (SCC-RI), Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Mickie Bhatia
- McMaster Stem Cell and Cancer Research Institute (SCC-RI), Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
50
|
Cuman C, Menkhorst E, Winship A, Van Sinderen M, Osianlis T, Rombauts LJ, Dimitriadis E. Fetal–maternal communication: the role of Notch signalling in embryo implantation. Reproduction 2014; 147:R75-86. [DOI: 10.1530/rep-13-0474] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The establishment of a successful pregnancy requires the implantation of a competent blastocyst into a ‘receptive’ endometrium, facilitating the formation of a functional placenta. Inadequate or inappropriate implantation and placentation is a major reason for infertility and is thought to lead to first-trimester miscarriage, placental insufficiency and other obstetric complications. Blastocyst–endometrial interactions are critical for implantation and placental formation. The Notch signalling family is a receptor–ligand family that regulates cellular processes as diverse as proliferation, apoptosis, differentiation, invasion and adhesion. Notch signalling is achieved via cell–cell interaction; thus, via Notch, cells can have direct effects on the fate of their neighbours. Recently, a number of studies have identified Notch receptors and ligands in the endometrium, blastocyst and placenta. This review collates current knowledge of this large receptor–ligand family and explores the role of Notch signalling during implantation and placentation, drawing on information from both human and animal studies. Overall, the evidence suggests that Notch signalling is a critical component of fetal–maternal communication during implantation and placentation and that abnormal Notch expression is associated with impaired placentation and pre-eclampsia.
Collapse
|