1
|
Li N, Zeng PYF, Kim HAJ, Karimi A, Ying S, Shaikh MH, Khan H, Joris K, Al Jawhri M, Cecchini M, Mymryk JS, Barrett JW, Nichols AC. Molecular features of T and N stage progression in laryngeal cancer. Oral Oncol 2025; 165:107283. [PMID: 40239580 DOI: 10.1016/j.oraloncology.2025.107283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/12/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Laryngeal squamous cell cancer (LSCC) is a common type of head and neck cancer that is typically unrelated to human papilloma virus (HPV) infection. Late-stage laryngeal cancers are associated with greater morbidity due to obstructive symptoms, and poorer overall survival. Using data from The Cancer Genome Atlas (TCGA), we analyzed 112 patient LSCC samples, comparing patient proteome, transcriptome and genome between early and late T and N samples. We observed significant differences in SNV frequency for various genes between the early and late-stage groups. Most notably we observed that NOTCH1 mutation, which was more frequent in late N-stage supraglottic cancers, was also associated with poorer patient survival in LSCCs. Methylation analysis also revealed changes in JUN gene methylation in late N glottic cancers. Transcriptomic analysis revealed differential expression in c-JUN, HOXB7 and HOXB9 transcript levels, suggesting potential involvement of these pathways in progression and nodal involvement. Our findings illustrate that LSCC undergoes distinct molecular changes associated with different stages and subsites. We observed multiple potential markers for progression, metastases and survival, including NOTCH1 mutation, which may aid as prognostic indicators in future studies.
Collapse
Affiliation(s)
- Nicholas Li
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Peter Y F Zeng
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Hugh A J Kim
- Department of Otolaryngology - Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Amir Karimi
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Shengjie Ying
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Mushfiq H Shaikh
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Halema Khan
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Krista Joris
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - MohdWessam Al Jawhri
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada
| | - Matthew Cecchini
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Joe S Mymryk
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada; Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada
| | - John W Barrett
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Anthony C Nichols
- Department of Otolaryngology - Head and Neck Surgery, University of Western Ontario, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
2
|
Magesh V, Sekar R, AlZahrani A, Balasubramanian R, Abdelsalam SA, Rajendran P. HES1 in cancer: a key player in tumorigenesis and its prognostic significance. Mol Genet Genomics 2025; 300:49. [PMID: 40392313 DOI: 10.1007/s00438-025-02259-1] [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: 02/09/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
The dysregulation of transcriptional regulators is a critical feature in the progression of many malignancies. Hairy and enhancer of split homolog-1 (HES1), a member of the basic helix-loop-helix (bHLH) gene family, has emerged as a key player in tumorigenesis due to its regulatory roles in multiple cellular pathways. This review aims to systematically explore the relevance of HES1 in cancer development, emphasizing its activation through major signaling pathways such as Notch, Hedgehog, hypoxia, and Wnt, and its contribution to advanced tumor progression. Numerous studies have demonstrated that HES1 upregulates genes associated with stemness, proliferation, and metastasis, and its expression correlates with poor clinicopathological features, including enhanced tumor proliferation, self-renewal, migration, metastasis, and drug resistance. Furthermore, HES1 has been frequently identified as a downstream effector of critical oncogenic pathways, further consolidating its role in aggressive cancers. Based on current evidence, HES1 holds promise as both a prognostic biomarker and a potential therapeutic target in various lethal malignancies. A deeper understanding of HES1's molecular mechanisms could pave the way for the development of targeted interventions aimed at improving cancer outcomes.
Collapse
Affiliation(s)
| | - Ramya Sekar
- Department of Oral & Maxillofacial Pathology and Oral Microbiology, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research (Deemed to Be University), Chennai, Tamil Nadu, India
| | - Abdullah AlZahrani
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | | | - Salaheldin Abdelraouf Abdelsalam
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt
| | - Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia.
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, Tamil Nadu, India.
| |
Collapse
|
3
|
Rodríguez-Lorca R, Román R, Beteta-Göbel R, Torres M, Lladó V, Escribá PV, Fernández-García P. Targeting the Notch-Furin axis with 2-hydroxyoleic acid: a key mechanism in glioblastoma therapy. Cell Oncol (Dordr) 2025; 48:373-390. [PMID: 39400678 PMCID: PMC11996967 DOI: 10.1007/s13402-024-00995-x] [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] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
PURPOSE Glioblastomas (GBMs) are highly treatment-resistant and aggressive brain tumors. 2OHOA, which is currently running a phase IIB/III clinical trial for newly diagnosed GBM patients, was developed in the context of melitherapy. This therapy focuses on the regulation of the membrane's structure and organization with the consequent modulation of certain cell signals to revert the pathological state in several disorders. Notch signaling has been associated with tumorigenesis and cell survival, potentially driving the pathogenesis of GBM. The current study aims to determine whether 2OHOA modulates the Notch pathway as part of its antitumoral mechanism. METHODS 2OHOA's effect was evaluated on different components of the pathway by Western blot, Q-PCR, and confocal microscopy. Notch receptor processing was analyzed by subcellular fractionation and colocalization studies. Furin activity was evaluated under cleavage of its substrate by fluorescence assays and its binding affinity to 2OHOA was determined by surface plasmon resonance. RESULTS We found that 2OHOA inhibits Notch2 and Notch3 signaling by dual mechanism. Notch2 inhibition is unleashed by impairment of its processing through the inactivation of furin activity by physical association. Instead, Notch3 is transcriptionally downregulated leading to a lower activation of the pathway. Moreover, we also found that HES1 overexpression highlighted the relevance of this pathway in the 2OHOA pharmacological efficacy. CONCLUSION These findings report that the inhibition of Notch signaling by 2OHOA plays a role in its anti-tumoral activity, an effect that may be driven through direct inhibition of furin, characterizing a novel target of this bioactive lipid to treat GBM.
Collapse
Affiliation(s)
- Raquel Rodríguez-Lorca
- Department of Biology, Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, Palma de Mallorca, 07122, Spain.
- R&D Department, Laminar Pharmaceuticals, C/Isaac Newton, Palma de Mallorca, 07121, Spain.
| | - Ramón Román
- Department of Biology, Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, Palma de Mallorca, 07122, Spain
- R&D Department, Laminar Pharmaceuticals, C/Isaac Newton, Palma de Mallorca, 07121, Spain
| | - Roberto Beteta-Göbel
- Department of Biology, Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, Palma de Mallorca, 07122, Spain
- R&D Department, Laminar Pharmaceuticals, C/Isaac Newton, Palma de Mallorca, 07121, Spain
| | - Manuel Torres
- Department of Biology, Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, Palma de Mallorca, 07122, Spain
| | - Victoria Lladó
- Department of Biology, Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, Palma de Mallorca, 07122, Spain
- R&D Department, Laminar Pharmaceuticals, C/Isaac Newton, Palma de Mallorca, 07121, Spain
| | - Pablo V Escribá
- Department of Biology, Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, Palma de Mallorca, 07122, Spain.
- R&D Department, Laminar Pharmaceuticals, C/Isaac Newton, Palma de Mallorca, 07121, Spain.
| | - Paula Fernández-García
- R&D Department, Laminar Pharmaceuticals, C/Isaac Newton, Palma de Mallorca, 07121, Spain
| |
Collapse
|
4
|
Agosti E, Antonietti S, Ius T, Fontanella MM, Zeppieri M, Panciani PP. Glioma Stem Cells as Promoter of Glioma Progression: A Systematic Review of Molecular Pathways and Targeted Therapies. Int J Mol Sci 2024; 25:7979. [PMID: 39063221 PMCID: PMC11276876 DOI: 10.3390/ijms25147979] [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: 05/30/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Gliomas' aggressive nature and resistance to therapy make them a major problem in oncology. Gliomas continue to have dismal prognoses despite significant advancements in medical science, and traditional treatments like surgery, radiation (RT), and chemotherapy (CT) frequently prove to be ineffective. After glioma stem cells (GSCs) were discovered, the traditional view of gliomas as homogeneous masses changed. GSCs are essential for tumor growth, treatment resistance, and recurrence. These cells' distinct capacities for differentiation and self-renewal are changing our knowledge of the biology of gliomas. This systematic literature review aims to uncover the molecular mechanisms driving glioma progression associated with GSCs. The systematic review adhered to PRISMA guidelines, with a thorough literature search conducted on PubMed, Ovid MED-LINE, and Ovid EMBASE. The first literature search was performed on 1 March 2024, and the search was updated on 15 May 2024. Employing MeSH terms and Boolean operators, the search focused on molecular mechanisms associated with GCSs-mediated glioma progression. Inclusion criteria encompassed English language studies, preclinical studies, and clinical trials. A number of 957 papers were initially identified, of which 65 studies spanning from 2005 to 2024 were finally included in the review. The main GSC model distribution is arranged in decreasing order of frequency: U87: 20 studies (32.0%); U251: 13 studies (20.0%); A172: 4 studies (6.2%); and T98G: 2 studies (3.17%). From most to least frequent, the distribution of the primary GSC pathway is as follows: Notch: 8 studies (12.3%); STAT3: 6 studies (9.2%); Wnt/β-catenin: 6 studies (9.2%); HIF: 5 studies (7.7%); and PI3K/AKT: 4 studies (6.2%). The distribution of molecular effects, from most to least common, is as follows: inhibition of differentiation: 22 studies (33.8%); increased proliferation: 18 studies (27.7%); enhanced invasive ability: 15 studies (23.1%); increased self-renewal: 5 studies (7.7%); and inhibition of apoptosis: 3 studies (4.6%). This work highlights GSC heterogeneity and the dynamic interplay within the glioblastoma microenvironment, underscoring the need for a tailored approach. A few key pathways influencing GSC behavior are JAK/STAT3, PI3K/AKT, Wnt/β-catenin, and Notch. Therapy may target these pathways. This research urges more study to fill in knowledge gaps in the biology of GSCs and translate findings into useful treatment approaches that could improve GBM patient outcomes.
Collapse
Affiliation(s)
- Edoardo Agosti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.); (P.P.P.)
| | - Sara Antonietti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.); (P.P.P.)
| | - Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Marco Maria Fontanella
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.); (P.P.P.)
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Pier Paolo Panciani
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.); (P.P.P.)
| |
Collapse
|
5
|
Wang S, Gu S, Chen J, Yuan Z, Liang P, Cui H. Mechanism of Notch Signaling Pathway in Malignant Progression of Glioblastoma and Targeted Therapy. Biomolecules 2024; 14:480. [PMID: 38672496 PMCID: PMC11048644 DOI: 10.3390/biom14040480] [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: 01/26/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of glioma and the most common primary tumor of the central nervous system. Despite significant advances in clinical management strategies and diagnostic techniques for GBM in recent years, it remains a fatal disease. The current standard of care includes surgery, radiation, and chemotherapy, but the five-year survival rate for patients is less than 5%. The search for a more precise diagnosis and earlier intervention remains a critical and urgent challenge in clinical practice. The Notch signaling pathway is a critical signaling system that has been extensively studied in the malignant progression of glioblastoma. This highly conserved signaling cascade is central to a variety of biological processes, including growth, proliferation, self-renewal, migration, apoptosis, and metabolism. In GBM, accumulating data suggest that the Notch signaling pathway is hyperactive and contributes to GBM initiation, progression, and treatment resistance. This review summarizes the biological functions and molecular mechanisms of the Notch signaling pathway in GBM, as well as some clinical advances targeting the Notch signaling pathway in cancer and glioblastoma, highlighting its potential as a focus for novel therapeutic strategies.
Collapse
Affiliation(s)
- Shenghao Wang
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China;
| | - Sikuan Gu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
| | - Junfan Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
| | - Zhiqiang Yuan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
| | - Ping Liang
- Department of Neurosurgery, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China;
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China; (S.G.); (J.C.); (Z.Y.)
- Department of Neurosurgery, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| |
Collapse
|
6
|
Li K, Hao W, Wu J, Liu X, Xing W, Zheng Y. Biofunctional study on chemoresistance in esophageal squamous carcinoma cells induced by missense mutation of NOTCH1 p.E450K. J Thorac Dis 2024; 16:1947-1959. [PMID: 38617785 PMCID: PMC11009606 DOI: 10.21037/jtd-23-1880] [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: 12/10/2023] [Accepted: 02/16/2024] [Indexed: 04/16/2024]
Abstract
Background Neoadjuvant chemotherapy (nCT) combined with surgery is one of the main strategies for the treatment of resectable locally advanced esophageal squamous cell carcinoma (ESCC). However, nearly 40% of patients did not benefit from nCT, and the detection rate of NOTCH1 missense mutation was significantly increased in patients who did not respond to chemotherapy, suggesting that the missense mutation may be related to tumor chemoresistance. We aim to explore the effect of a NOTCH1 missense mutation on cell phenotype, to interpret the biofunctional changes in cell lines with a NOTCH1 missense mutation and to analyze the effect of a NOTCH1 missense mutation on drug resistance in ESCC cell lines. Methods Sanger sequencing was used to evaluate the exon mutations in the NOTCH1 ligand binding region of candidate ESCC cell lines. After screening, KYSE450 and KYSE140 cells were selected as the research objects, and point mutation cell lines [KYSE140-mutant-type (MT) and KYSE450-MT] were constructed by CRISPR/Cas9 technology. Then, functional experiments were performed with the four cell lines [KYSE450-MT/wild-type (WT) and KYSE140-MT/WT]. The drug resistance of ESCC cell lines was assessed with a drug sensitivity test, and the proliferation, invasion and migration of ESCC lines were evaluated by proliferation test, scratch test and Transwell test. The cell cycle status of ESCC cells was assessed using flow cytometry. Results Drug sensitivity tests showed that the NOTCH1 p.E450K point mutation caused chemotherapy resistance in KYSE140 and KYSE450 ESCC cell lines. Cell proliferation, Wound scratch and Transwell assays showed that the NOTCH1 p.E450K point mutation enhanced the proliferation, invasion and migration abilities of KYSE140 and KYSE450 cells. Flow cytometry analysis showed that the NOTCH1 p.E450K point mutation caused an increase in KYSE140 and KYSE450 cells in S phase. Conclusions The NOTCH1 p.E450K point mutation causes chemotherapy resistance in KYSE140 and KYSE450 ESCC cells. Cell functional experiments showed that the NOTCH1 p.E450K point mutation enhanced the proliferation, migration and invasion abilities of KYSE140 and KYSE450 cells and increased the number of cells in S phase.
Collapse
Affiliation(s)
- Keting Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou, China
| | - Wentao Hao
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Shanghai, China
| | - Jiwei Wu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou, China
| | - Xianben Liu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou, China
| | - Wenqun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou, China
| | - Yan Zheng
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
7
|
Shahcheraghi SH, Asl ER, Lotfi M, Ayatollahi J, Khaleghinejad SH, Aljabali AAA, Bakshi HA, El-Tanani M, Charbe NB, Serrano-Aroca Á, Mishra V, Mishra Y, Goyal R, Hromić-Jahjefendić A, Uversky VN, Lotfi M, Tambuwala MM. Non-coding RNAs as Key Regulators of the Notch Signaling Pathway in Glioblastoma: Diagnostic, Prognostic, and Therapeutic Targets. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1203-1216. [PMID: 38279763 DOI: 10.2174/0118715273277458231213063147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 01/28/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly invasive brain malignancy originating from astrocytes, accounting for approximately 30% of central nervous system malignancies. Despite advancements in therapeutic strategies including surgery, chemotherapy, and radiopharmaceutical drugs, the prognosis for GBM patients remains dismal. The aggressive nature of GBM necessitates the identification of molecular targets and the exploration of effective treatments to inhibit its proliferation. The Notch signaling pathway, which plays a critical role in cellular homeostasis, becomes deregulated in GBM, leading to increased expression of pathway target genes such as MYC, Hes1, and Hey1, thereby promoting cellular proliferation and differentiation. Recent research has highlighted the regulatory role of non-coding RNAs (ncRNAs) in modulating Notch signaling by targeting critical mRNA expression at the post-transcriptional or transcriptional levels. Specifically, various types of ncRNAs, including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), have been shown to control multiple target genes and significantly contribute to the carcinogenesis of GBM. Furthermore, these ncRNAs hold promise as prognostic and predictive markers for GBM. This review aims to summarize the latest studies investigating the regulatory effects of ncRNAs on the Notch signaling pathway in GBM.
Collapse
Affiliation(s)
- Seyed Hossein Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elmira Roshani Asl
- Social Determinants of Health Research Center, Saveh University of Medical Sciences, Saveh, Iran
| | - Malihe Lotfi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshid Ayatollahi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Hematology and Oncology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Hamid A Bakshi
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Mohamed El-Tanani
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Nitin B Charbe
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA
| | - Ángel Serrano-Aroca
- Biomaterials & Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, San Vicente Mártir, Valencia, 46001, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan, India
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, 71000 Sarajevo, Bosnia and Herzegovina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Marzieh Lotfi
- Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
| |
Collapse
|
8
|
Srinivas N, Song L, Lei KC, Gravemeyer J, Furtmann F, Gambichler T, Becker JC, Sriram A. The HDAC inhibitor domatinostat induces type I interferon α in Merkel cell carcinoma by HES1 repression. J Cancer Res Clin Oncol 2023; 149:8267-8277. [PMID: 37071208 PMCID: PMC10374800 DOI: 10.1007/s00432-023-04733-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Class I selective histone deacetylase inhibitors (HDACi) have been previously demonstrated to not only increase major histocompatibility complex class I surface expression in Merkel cell carcinoma (MCC) cells by restoring the antigen processing and presentation machinery, but also exert anti-tumoral effect by inducing apoptosis. Both phenomena could be due to induction of type I interferons (IFN), as has been described for HDACi. However, the mechanism of IFN induction under HDACi is not fully understood because the expression of IFNs is regulated by both activating and inhibitory signaling pathways. Our own preliminary observations suggest that this may be caused by suppression of HES1. METHODS The effect of the class I selective HDACi domatinostat and IFNα on cell viability and the apoptosis of MCPyV-positive (WaGa, MKL-1) and -negative (UM-MCC 34) MCC cell lines, as well as, primary fibroblasts were assessed by colorimetric methods or measuring mitochondrial membrane potential and intracellular caspase-3/7, respectively. Next, the impact of domatinostat on IFNA and HES1 mRNA expression was measured by RT-qPCR; intracellular IFNα production was detected by flow cytometry. To confirm that the expression of IFNα induced by HDACi was due to the suppression of HES1, it was silenced by RNA interference and then mRNA expression of IFNA and IFN-stimulated genes was assessed. RESULTS Our studies show that the previously reported reduction in viability of MCC cell lines after inhibition of HDAC by domatinostat is accompanied by an increase in IFNα expression, both of mRNA and at the protein level. We confirmed that treatment of MCC cells with external IFNα inhibited their proliferation and induced apoptosis. Re-analysis of existing single-cell RNA sequencing data indicated that induction of IFNα by domatinostat occurs through repression of HES1, a transcriptional inhibitor of IFNA; this was confirmed by RT-qPCR. Finally, siRNA-mediated silencing of HES1 in the MCC cell line WaGa not only increased mRNA expression of IFNA and IFN-stimulated genes but also decreased cell viability. CONCLUSION Our results demonstrate that the direct anti-tumor effect of HDACi domatinostat on MCC cells is at least in part mediated via decreased HES1 expression allowing the induction of IFNα, which in turn causes apoptosis.
Collapse
Affiliation(s)
- Nalini Srinivas
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Lina Song
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kuan Cheok Lei
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Gravemeyer
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frauke Furtmann
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thilo Gambichler
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Jürgen C. Becker
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Ashwin Sriram
- Department of Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), Partner Site Essen, University Medicine Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
9
|
Cao Y, Liu B, Cai L, Li Y, Huang Y, Zhou Y, Sun X, Yang W, Sun T. G9a promotes immune suppression by targeting the Fbxw7/Notch pathway in glioma stem cells. CNS Neurosci Ther 2023; 29:2508-2521. [PMID: 36971192 PMCID: PMC10401078 DOI: 10.1111/cns.14191] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/22/2023] [Accepted: 03/09/2023] [Indexed: 08/05/2023] Open
Abstract
AIM Immunotherapy for glioblastoma multiforme (GBM) is limited because of a strongly immunosuppressive tumor microenvironment (TME). Remodeling the immune TME is an effective strategy to eliminate GBM immunotherapy resistance. Glioma stem cells (GSCs) are inherently resistant to chemotherapy and radiotherapy and involved in immune evasion mechanism. This study aimed to investigate the effects of histone methyltransferases 2 (EHMT2 or G9a) on immunosuppressive TME and whether this effect was related to changes on cell stemness. METHODS Tumor-infiltrating immune cells were analyzed by flow cytometry and immunohistochemistry in orthotopic implanted glioma mice model. The gene expressions were measured by RT-qPCR, western blot, immunofluorescence, and flow cytometry. Cell viability was detected by CCK-8, and cell apoptosis and cytotoxicity were detected by flow cytometry. The interaction of G9a and F-box and WD repeat domain containing 7 (Fbxw7) promotor was verified by dual-luciferase reporter assay and chromatin immunoprecipitation. RESULTS Downregulation of G9a retarded tumor growth and extended survival in an immunocompetent glioma mouse model, promoted the filtration of IFN-γ + CD4+ and CD8+ T lymphocytes, and suppressed the filtration of PD-1+ CD4+ and CD8+ T lymphocytes, myeloid-derived suppressor cells (MDSCs) and M2-like macrophages in TME. G9a inhibition decreased PD-L1 and increased MHC-I expressions by inactivating Notch pathway companying stemness decrease in GSCs. Mechanistically, G9a bound to Fbxw7, a Notch suppressor, to inhibit gene transcription through H3K9me2 of Fbxw7 promotor. CONCLUSION G9a promotes stemness characteristics through binding Fbxw7 promotor to inhibit Fbxw7 transcription in GSCs, forming an immunosuppressive TME, which provides novel treatment strategies for targeting GSCs in antitumor immunotherapy.
Collapse
Affiliation(s)
- Yufei Cao
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bin Liu
- Department of Neurosurgery, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Lize Cai
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanyan Li
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yulun Huang
- Department of Neurosurgery, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, Jiangsu, China
| | - Youxin Zhou
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xingjian Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China
| | - Wei Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China
| | - Ting Sun
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
10
|
Wei L, Pan M, Jiang Q, Hu B, Zhao J, Zou C, Chen L, Tang C, Zou D. Eukaryotic initiation factor 4 A-3 promotes glioblastoma growth and invasion through the Notch1-dependent pathway. BMC Cancer 2023; 23:550. [PMID: 37322413 DOI: 10.1186/s12885-023-10946-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND As an adult tumor with the most invasion and the highest mortality rate, the inherent heterogeneity of glioblastoma (GBM) is the main factor that causes treatment failure. Therefore, it is important to have a deeper understanding of the pathology of GBM. Some studies have shown that Eukaryotic Initiation Factor 4A-3 (EIF4A3) can promote the growth of many people's tumors, and the role of specific molecules in GBM remains unclear. METHODS The correlation between the expression of EIF4A3 gene and its prognosis was studied in 94 GBM patients using survival analysis. Further in vitro and in vivo experiments, the effect of EIF4A3 on GBM cells proliferation, migration, and the mechanism of EIF4A3 on GBM was explored. In addition, combined with bioinformatics analysis, we further confirmed that EIF4A3 contributes to the progress of GBM. RESULTS The expression of EIF4A3 was upregulated in GBM tissues, and high expression of EIF4A3 is associated with poor prognosis in GBM. In vitro, knockdown of EIF4A3 significantly reduced the proliferation, migration, and invasion abilities of GBM cells, whereas overexpression of EIF4A3 led to the opposite effect. The analysis of differentially expressed genes related to EIF4A3 indicates that it is involved in many cancer-related pathways, such as Notch and JAK-STAT3 signal pathway. In Besides, we demonstrated the interaction between EIF4A3 and Notch1 by RNA immunoprecipitation. Finally, the biological function of EIF4A3-promoted GBM was confirmed in living organisms. CONCLUSION The results of this study suggest that EIF4A3 may be a potential prognostic factor, and Notch1 participates in the proliferation and metastasis of GBM cells mediated by EIF4A3.
Collapse
Affiliation(s)
- Lei Wei
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Qiulan Jiang
- Department of Radiation Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, People's Republic of China
| | - Beiquan Hu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, China
| | - Jianyi Zhao
- Department of Neurosurgery, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Chun Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Liechun Chen
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Chunhai Tang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
- The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue Dong Road, Nanning, 530007, Guangxi, China.
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
- The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue Dong Road, Nanning, 530007, Guangxi, China.
| |
Collapse
|
11
|
Paul M, Chakraborty S, Islam S, Ain R. Trans-differentiation of trophoblast stem cells: implications in placental biology. Life Sci Alliance 2023; 6:6/3/e202201583. [PMID: 36574992 PMCID: PMC9797987 DOI: 10.26508/lsa.202201583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
Trophoblast invasion is a hallmark of hemochorial placentation. Invasive trophoblast cells replace the endothelial cells of uterine spiral arteries. The mechanism by which the invasive trophoblast cells acquire this phenotype is unknown. Here, we demonstrate that, during differentiation, a small population of trophoblast stem (TS) cells trans-differentiate into a hybrid cell type expressing markers of both trophoblast (TC) and endothelial (EC) cells. In addition, a compendium of EC-specific genes was found to be associated with TS cell differentiation. Using functional annotation, these genes were categorized into angiogenesis, cell adhesion molecules, and apoptosis-related genes. HES1 repressed transcription of EC genes in TS cells. Interestingly, differentiated TCs secrete TRAIL, but its receptor DR4 is expressed only in ECs and not in TCs. TRAIL induced apoptosis in EC but not in TC. Co-culture of ECs with TC induced apoptosis in ECs via extrinsic apoptotic pathway. These results highlight that (a) TS cells possess the potential to trans-differentiate into "trophendothelial" phenotype, regulated by HES1 and (b) trophoblast differentiation-induced TRAIL secretion directs preferential demise of ECs located in their vicinity.
Collapse
Affiliation(s)
- Madhurima Paul
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Shreeta Chakraborty
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,National Institutes of Health, Bethesda, MD, USA
| | - Safirul Islam
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,School of Biotechnology, Presidency University, Kolkata, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
12
|
Kurtović M, Piteša N, Bartoniček N, Ozretić P, Musani V, Čonkaš J, Petrić T, King C, Sabol M. RNA-seq and ChIP-seq Identification of Unique and Overlapping Targets of GLI Transcription Factors in Melanoma Cell Lines. Cancers (Basel) 2022; 14:4540. [PMID: 36139698 PMCID: PMC9497141 DOI: 10.3390/cancers14184540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite significant progress in therapy, melanoma still has a rising incidence worldwide, and novel treatment strategies are needed. Recently, researchers have recognized the involvement of the Hedgehog-GLI (HH-GLI) signaling pathway in melanoma and its consistent crosstalk with the MAPK pathway. In order to further investigate the link between the two pathways and to find new target genes that could be considered for combination therapy, we set out to find transcriptional targets of all three GLI proteins in melanoma. METHODS We performed RNA sequencing on three melanoma cell lines (CHL-1, A375, and MEL224) with overexpressed GLI1, GLI2, and GLI3 and combined them with the results of ChIP-sequencing on endogenous GLI1, GLI2, and GLI3 proteins. After combining these results, 21 targets were selected for validation by qPCR. RESULTS RNA-seq revealed a total of 808 differentially expressed genes (DEGs) for GLI1, 941 DEGs for GLI2, and 58 DEGs for GLI3. ChIP-seq identified 527 genes that contained GLI1 binding sites in their promoters, 1103 for GLI2 and 553 for GLI3. A total of 15 of these targets were validated in the tested cell lines, 6 of which were detected by both RNA-seq and ChIP-seq. CONCLUSIONS Our study provides insight into the unique and overlapping transcriptional output of the GLI proteins in melanoma. We suggest that our findings could provide new potential targets to consider while designing melanoma-targeted therapy.
Collapse
Affiliation(s)
- Matea Kurtović
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Nikolina Piteša
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Nenad Bartoniček
- The Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
- The Kinghorn Centre for Clinical Genomics, 370 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Petar Ozretić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Vesna Musani
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Josipa Čonkaš
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Tina Petrić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Cecile King
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maja Sabol
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| |
Collapse
|
13
|
Dome A, Dymova M, Richter V, Stepanov G. Post-Transcriptional Modifications of RNA as Regulators of Apoptosis in Glioblastoma. Int J Mol Sci 2022; 23:9272. [PMID: 36012529 PMCID: PMC9408889 DOI: 10.3390/ijms23169272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
This review is devoted to changes in the post-transcriptional maturation of RNA in human glioblastoma cells, which leads to disruption of the normal course of apoptosis in them. The review thoroughly highlights the latest information on both post-transcriptional modifications of certain regulatory RNAs, associated with the process of apoptosis, presents data on the features of apoptosis in glioblastoma cells, and shows the relationship between regulatory RNAs and the apoptosis in tumor cells. In conclusion, potential target candidates are presented that are necessary for the development of new drugs for the treatment of glioblastoma.
Collapse
Affiliation(s)
| | - Maya Dymova
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | | | | |
Collapse
|
14
|
Cifola I, Fratini F, Cardinali B, Palmieri V, Gatti G, Selmi T, Donzelli S, Sacconi A, Cesarini V, Marei HE, Papi M, Blandino G, Cenciarelli C, Falcone G, D’Agnano I. miRNome and Proteome Profiling of Small Extracellular Vesicles Secreted by Human Glioblastoma Cell Lines and Primary Cancer Stem Cells. Biomedicines 2022; 10:1886. [PMID: 36009432 PMCID: PMC9405730 DOI: 10.3390/biomedicines10081886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/02/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor in adults. Despite available therapeutic interventions, it is very difficult to treat, and a cure is not yet available. The intra-tumoral GBM heterogeneity is a crucial factor contributing to poor clinical outcomes. GBM derives from a small heterogeneous population of cancer stem cells (CSCs). In cancer tissue, CSCs are concentrated within the so-called niches, where they progress from a slowly proliferating phase. CSCs, as most tumor cells, release extracellular vesicles (EVs) into the surrounding microenvironment. To explore the role of EVs in CSCs and GBM tumor cells, we investigated the miRNA and protein content of the small EVs (sEVs) secreted by two GBM-established cell lines and by GBM primary CSCs using omics analysis. Our data indicate that GBM-sEVs are selectively enriched for miRNAs that are known to display tumor suppressor activity, while their protein cargo is enriched for oncoproteins and tumor-associated proteins. Conversely, among the most up-regulated miRNAs in CSC-sEVs, we also found pro-tumor miRNAs and proteins related to stemness, cell proliferation, and apoptosis. Collectively, our findings support the hypothesis that sEVs selectively incorporate different miRNAs and proteins belonging both to fundamental processes (e.g., cell proliferation, cell death, stemness) as well as to more specialized ones (e.g., EMT, membrane docking, cell junction organization, ncRNA processing).
Collapse
Affiliation(s)
- Ingrid Cifola
- Institute for Biomedical Technologies (ITB), CNR, 20054 Segrate, Italy
| | - Federica Fratini
- Proteomics Core Facility, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Beatrice Cardinali
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy
| | - Valentina Palmieri
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
- Dipartimento di Neuroscienze, Sez. Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Institute for Complex Systems (ISC), CNR, 00185 Rome, Italy
| | - Giuliana Gatti
- Department of Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Tommaso Selmi
- Institute for Biomedical Technologies (ITB), CNR, 20054 Segrate, Italy
| | - Sara Donzelli
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Andrea Sacconi
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | | | - Hany E. Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt
| | - Massimilano Papi
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
- Dipartimento di Neuroscienze, Sez. Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology (IFT), CNR, 00133 Rome, Italy
| | - Germana Falcone
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 00015 Monterotondo, Italy
| | - Igea D’Agnano
- Institute for Biomedical Technologies (ITB), CNR, 20054 Segrate, Italy
| |
Collapse
|
15
|
Bilateral Feedback in Oscillator Model Is Required to Explain the Coupling Dynamics of Hes1 with the Cell Cycle. MATHEMATICS 2022. [DOI: 10.3390/math10132323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Biological processes are governed by the expression of proteins, and for some proteins, their level of expression can fluctuate periodically over time (i.e., they oscillate). Many oscillatory proteins (e.g., cell cycle proteins and those from the HES family of transcription factors) are connected in complex ways, often within large networks. This complexity can be elucidated by developing intuitive mathematical models that describe the underlying critical aspects of the relationships between these processes. Here, we provide a mathematical explanation of a recently discovered biological phenomenon: the phasic position of the gene Hes1’s oscillatory expression at the beginning of the cell cycle of an individual human breast cancer stem cell can have a predictive value on how long that cell will take to complete a cell cycle. We use a two-component model of coupled oscillators to represent Hes1 and the cell cycle in the same cell with minimal assumptions. Inputting only the initial phase angles, we show that this model is capable of predicting the dynamic mitosis to mitosis behaviour of Hes1 and predicting cell cycle length patterns as found in real-world experimental data. Moreover, we discover that bidirectional coupling between Hes1 and the cell cycle is critical within the system for the data to be reproduced and that nonfixed asymmetry in the interactions between the oscillators is required. The phase dynamics we present here capture the complex interplay between Hes1 and the cell cycle, helping to explain nongenetic cell cycle variability, which has critical implications in cancer treatment contexts.
Collapse
|
16
|
Notch signaling in malignant gliomas: supporting tumor growth and the vascular environment. Cancer Metastasis Rev 2022; 41:737-747. [PMID: 35624227 DOI: 10.1007/s10555-022-10041-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/18/2022] [Indexed: 11/02/2022]
Abstract
Glioblastoma is the most malignant form of glioma, which is the most commonly occurring tumor of the central nervous system. Notch signaling in glioblastoma is considered to be a marker of an undifferentiated tumor cell state, associated with tumor stem cells. Notch is also known for facilitating tumor dormancy escape, recurrence and progression after treatment. Studies in vitro suggest that reducing, removing or blocking the expression of this gene triggers tumor cell differentiation, which shifts the phenotype away from stemness status and consequently facilitates treatment. In contrast, in the vasculature, Notch appears to also function as an important receptor that defines mature non-leaking vessels, and increasing its expression promotes tumor normalization in models of cancer in vivo. Failures in clinical trials with Notch inhibitors are potentially related to their opposing effects on the tumor versus the tumor vasculature, which points to the need for a greater understanding of this signaling pathway.
Collapse
|
17
|
Shafi O, Siddiqui G. Tracing the origins of glioblastoma by investigating the role of gliogenic and related neurogenic genes/signaling pathways in GBM development: a systematic review. World J Surg Oncol 2022; 20:146. [PMID: 35538578 PMCID: PMC9087910 DOI: 10.1186/s12957-022-02602-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/15/2022] [Indexed: 02/16/2023] Open
Abstract
Background Glioblastoma is one of the most aggressive tumors. The etiology and the factors determining its onset are not yet entirely known. This study investigates the origins of GBM, and for this purpose, it focuses primarily on developmental gliogenic processes. It also focuses on the impact of the related neurogenic developmental processes in glioblastoma oncogenesis. It also addresses why glial cells are at more risk of tumor development compared to neurons. Methods Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving glioblastoma, gliogenesis, neurogenesis, stemness, neural stem cells, gliogenic signaling and pathways, neurogenic signaling and pathways, and astrocytogenic genes. Results The origin of GBM is dependent on dysregulation in multiple genes and pathways that accumulatively converge the cells towards oncogenesis. There are multiple layers of steps in glioblastoma oncogenesis including the failure of cell fate-specific genes to keep the cells differentiated in their specific cell types such as p300, BMP, HOPX, and NRSF/REST. There are genes and signaling pathways that are involved in differentiation and also contribute to GBM such as FGFR3, JAK-STAT, and hey1. The genes that contribute to differentiation processes but also contribute to stemness in GBM include notch, Sox9, Sox4, c-myc gene overrides p300, and then GFAP, leading to upregulation of nestin, SHH, NF-κB, and others. GBM mutations pathologically impact the cell circuitry such as the interaction between Sox2 and JAK-STAT pathway, resulting in GBM development and progression. Conclusion Glioblastoma originates when the gene expression of key gliogenic genes and signaling pathways become dysregulated. This study identifies key gliogenic genes having the ability to control oncogenesis in glioblastoma cells, including p300, BMP, PAX6, HOPX, NRSF/REST, LIF, and TGF beta. It also identifies key neurogenic genes having the ability to control oncogenesis including PAX6, neurogenins including Ngn1, NeuroD1, NeuroD4, Numb, NKX6-1 Ebf, Myt1, and ASCL1. This study also postulates how aging contributes to the onset of glioblastoma by dysregulating the gene expression of NF-κB, REST/NRSF, ERK, AKT, EGFR, and others.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan.
| | - Ghazia Siddiqui
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
18
|
Wang Y, Cheng Y, Yang Q, Kuang L, Liu G. Overexpression of FOXD2-AS1 enhances proliferation and impairs differentiation of glioma stem cells by activating the NOTCH pathway via TAF-1. J Cell Mol Med 2022; 26:2620-2632. [PMID: 35419917 PMCID: PMC9077300 DOI: 10.1111/jcmm.17268] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 02/20/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Emerging data have highlighted the importance of long noncoding RNAs (lncRNAs) in exerting critical biological functions and roles in different forms of brain cancer, including gliomas. In this study, we sought to investigate the role of lncRNA FOXD2 adjacent opposite strand RNA 1 (FOXD2-AS1) in glioma cells. First, we used sphere formation assay and flow cytometry to select U251 glioma stem cells (GSCs). Then, we quantified the expression of lncRNA FOXD2-AS1, TATA-box binding protein associated factor 1 (TAF-1) and NOTCH1 in glioma tissues and GSCs, as well as the expression of GSC stem markers, OCT4, SOX2, Nanog, Nestin and CD133 in GSCs. Colony formation assay, sphere formation assay, and flow cytometry were used to evaluate GSC stemness. Next, the correlations among lncRNA FOXD2-AS1, TAF-1 and NOTCH1 were investigated. LncRNA FOXD2-AS1, TAF-1 and NOTCH1 were found to be elevated in glioma tissues and GSCs, and silencing lncRNA FOXD2-AS1 inhibited stemness and proliferation, while promoting apoptosis and differentiation of GSCs. LncRNA FOXD2-AS1 overexpression also led to increased NOTCH1 by recruiting TAF-1 to the NOTCH1 promoter region, thereby promoting stemness and proliferation, while impairing cell apoptosis and differentiation. Mechanistically, lncRNA FOXD2-AS1 elevation promoted glioma in vivo by activating the NOTCH signalling pathway via TAF-1 upregulation. Taken together, the key findings of our investigation support the proposition that downregulation of lncRNA FOXD2-AS1 presents a viable and novel molecular candidate for improving glioma treatment.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurotumor Disease Treatment Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yanli Cheng
- Department of Dermatology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Qi Yang
- Department of Orthopeadic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Lei Kuang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Guolei Liu
- Department of Otorhinolaryngology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
19
|
Marei HE, Althani A, Afifi N, Hasan A, Caceci T, Felsani A, Tringali G, Cifola I, Pozzoli G, Cenciarelli C. Exome sequencing of glioblastoma-derived cancer stem cells reveals rare clinically relevant frameshift deletion in MLLT1 gene. Cancer Cell Int 2022; 22:9. [PMID: 34996478 PMCID: PMC8740446 DOI: 10.1186/s12935-021-02419-4] [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: 07/30/2021] [Accepted: 12/19/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a heterogeneous CNS neoplasm which causes significant morbidity and mortality. One reason for the poor prognostic outcome of GBM is attributed to the presence of cancer stem cells (CSC) which confer resistance against standard chemo- and radiotherapeutics modalities. Two types of GBM-associated CSC were isolated from the same patient: tumor core- (c-CSC) and peritumor tissue-derived cancer stem cells (p-CSC). Our experiments are focused on glioblastoma-IDH-wild type, and no disease-defining alterations were present in histone, BRAF or other genes. METHODS In the present study, potential differences in genetic variants between c-CSC versus p-CSC derived from four GBM patients were investigated with the aims of (1) comparing the exome sequences between all the c-CSC or p-CSC to identify the common variants; (2) identifying the variants affecting the function of genes known to be involved in cancer origin and development. RESULTS By comparative analyses, we identified common gene single nucleotide variants (SNV) in all GBM c-CSC and p-CSC, a potentially deleterious variant was a frameshift deletion at Gln461fs in the MLLT1 gene, that was encountered only in p-CSC samples with different allelic frequency. CONCLUSIONS We discovered a potentially harmful frameshift deletion at Gln461fs in the MLLT1 gene. Further investigation is required to confirm the presence of the identified mutations in patient tissue samples, as well as the significance of the frameshift mutation in the MLLT1 gene on GBM biology and response to therapy based on genomic functional experiments.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Asmaa Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Thomas Caceci
- Biomedical Sciences, Virginia Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Armando Felsani
- Genomnia S.R.L, Via Ludovico Ariosto, Bresso, MI, 20091, USA
| | - Giuseppe Tringali
- Institute of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), Milan, Italy
| | - Giacomo Pozzoli
- Pharmacology Unit, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere, 100, 00133, Rome, Italy
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere, 100, 00133, Rome, Italy.
| |
Collapse
|
20
|
Differential phase register of Hes1 oscillations with mitoses underlies cell-cycle heterogeneity in ER + breast cancer cells. Proc Natl Acad Sci U S A 2021; 118:2113527118. [PMID: 34725165 PMCID: PMC8609326 DOI: 10.1073/pnas.2113527118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Tumors exhibit heterogeneities that are not due to mutations, including cancer stem cells with different potencies. We show that the cancer stem-cell state predisposed to dormancy in vivo has a highly variable and long cell cycle. Using single-cell live imaging for the transcriptional repressor Hes1 (a key molecule in cancer), we show a type of circadian-like oscillatory expression of Hes1 in all cells in the population. The most potent cancer stem cells tend to divide around the trough of the Hes1 oscillatory wave, a feature predictive of a long cell cycle. A concept proposed here is that the position of cell division with respect to the Hes1 wave is predictive of its prospective cell-cycle length and cancer cellular substate. Here, we study the dynamical expression of endogenously labeled Hes1, a transcriptional repressor implicated in controlling cell proliferation, to understand how cell-cycle length heterogeneity is generated in estrogen receptor (ER)+ breast cancer cells. We find that Hes1 shows oscillatory expression with ∼25 h periodicity and during each cell cycle has a variable peak in G1, a trough around G1–S transition, and a less variable second peak in G2/M. Compared to other subpopulations, the cell cycle in CD44HighCD24Low cancer stem cells is longest and most variable. Most cells divide around the peak of the Hes1 expression wave, but preceding mitoses in slow dividing CD44HighCD24Low cells appear phase-shifted, resulting in a late-onset Hes1 peak in G1. The position, duration, and shape of this peak, rather than the Hes1 expression levels, are good predictors of cell-cycle length. Diminishing Hes1 oscillations by enforcing sustained expression slows down the cell cycle, impairs proliferation, abolishes the dynamic expression of p21, and increases the percentage of CD44HighCD24Low cells. Reciprocally, blocking the cell cycle causes an elongation of Hes1 periodicity, suggesting a bidirectional interaction of the Hes1 oscillator and the cell cycle. We propose that Hes1 oscillations are functionally important for the efficient progression of the cell cycle and that the position of mitosis in relation to the Hes1 wave underlies cell-cycle length heterogeneity in cancer cell subpopulations.
Collapse
|
21
|
Zhu Y, Wang H, Fei M, Tang T, Niu W, Zhang L. Smarcd1 Inhibits the Malignant Phenotypes of Human Glioblastoma Cells via Crosstalk with Notch1. Mol Neurobiol 2021; 58:1438-1452. [PMID: 33190170 PMCID: PMC7932991 DOI: 10.1007/s12035-020-02190-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 10/29/2020] [Indexed: 11/29/2022]
Abstract
Smarcd1 is a component of an evolutionary conserved chromatin remodeling complex-SWI/SNF, which is involved in transcription factor recruitment, DNA replication, recombination, and repair. Suppression of the SWI/SNF complex required for cellular differentiation and gene regulation may be inducible for cell proliferation and tumorigenicity. However, the inhibitory role of Smarcd1 in human glioblastoma cells has not been well illustrated. Both U87 and U251 human glioblastoma cell lines were employed in the present study. The lentivirus-mediated gene knockdown and overexpression approach was conducted to determine the function of Smarcd1. The protein levels were tested by western blot, and the relative mRNA contents were detected by quantitative real-time PCR. Cell viability was tested by CCK-8 and colony-forming assay. Transwell assays were utilized to evaluate the motility and invasive ability. Flow cytometry was employed to analyze cell cycle and apoptosis. SPSS software was used for statistical analysis. Low expression of Smarcd1 was observed in glioblastoma cell lines and in patients with high-grade glioma. Importantly, the depletion of Smarcd1 promoted cell proliferation, invasion, and chemoresistance, whereas enhanced expression of Smarcd1 inhibited tumor-malignant phenotypes. Mechanistic research demonstrated that overexpression of Smarcd1 decreased the expression of Notch1, while knockdown of Notch1 increased the expression of Smarcd1 through Hes1 suppression. Hence, the crosstalk between Smarcd1 and Notch1, which formed a feedback loop, was crucial in regulation of glioblastoma malignant phenotypes. Furthermore, targeting Smarcd1 could be a potential strategy for human glioblastoma treatment.
Collapse
Affiliation(s)
- Yihao Zhu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China.
| | - Maoxing Fei
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Ting Tang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Wenhao Niu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Li Zhang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| |
Collapse
|
22
|
Ghosh S, Khanam R, Acharya Chowdhury A. The Evolving Roles of Bacopa monnieri as Potential Anti-Cancer Agent: A Review. Nutr Cancer 2020; 73:2166-2176. [PMID: 33148034 DOI: 10.1080/01635581.2020.1841248] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The intermingled interrelationship of Bacopa monnieri and human health dates backs to the ancient times in the history of ayurveda making the plant an enriched source of alternative drug development in a nontoxic manner. In recent years, research on the biological effects of Bacopa monnieri has flourished as promising neuroprotective, memory boosting and more importantly as both chemopreventive and anti-neoplastic agent. Each naturally synthesized chemical constituent identified from Bacopa monnieri leaf extract with different solvents, has significant anti-metastatic, anti-angiogenic and anti-proliferative activity on different type of cancer cells. In this context, a substantial literature survey allows a deep understanding of the involvement of specific bioactive molecules along with the whole plant extract of Bacopa monnieri with their divergent effective molecular pathways. This comprehensive review covers literature up to the year 2020 highlighting all the anticancer efficacy along with signaling pathways activated by secondary metabolites found in bacopa plant.
Collapse
Affiliation(s)
- Sudeepa Ghosh
- Department of Biotechnology, JIS University, Kolkata, West Bengal, India
| | - Rahmat Khanam
- Department of Biotechnology, JIS University, Kolkata, West Bengal, India
| | | |
Collapse
|
23
|
Sun Z, Wang L, Zhou Y, Dong L, Ma W, Lv L, Zhang J, Wang X. Glioblastoma Stem Cell-Derived Exosomes Enhance Stemness and Tumorigenicity of Glioma Cells by Transferring Notch1 Protein. Cell Mol Neurobiol 2020; 40:767-784. [PMID: 31853695 PMCID: PMC11448788 DOI: 10.1007/s10571-019-00771-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/03/2019] [Indexed: 02/05/2023]
Abstract
Exosomes contain plenty of bioactive information, playing an important role in intercellular communication by transfer their bioactive molecular contents to recipient cells. Glioblastoma stem cells (GSCs) and non-GSC glioma cells coexist in GBM microenvironment; GSC-released exosomes contain intracellular signaling molecules, which may affect the biological phenotypes of recipient cells. However, whether GSC exosomes could affect the biological phenotype of non-GSC glioma cells has not yet been defined. To explore whether GSC exosomes could reprogramme non-GSC glioma cells into GSCs and its possible mechanism involved, non-GSC glioma cells were treated with GSCs released exosomes; the potential mechanisms of action were studied with RNA interference, Notch inhibitors and Western blot analysis. The proliferation, neurosphere formation, invasive capacities, and tumorigenicity of non-GSC glioma cells were increased significantly after GSC exosome treatment; Notch1 signaling pathway was activated in GSCs; Notch1 protein was highly enriched in GSC exosomes; Notch1 signaling pathway and stemness-related protein expressions were increased in GSC exosome treated non-GSC glioma cells and these cell generated tumor tissues; Notch1 protein expression in GSCs and their exosomes, and the neurosphere formation of GSCs were decreased by Notch1 RNA interference; Notch1 signaling pathway protein and stemness protein expressions were decreased in GSC exosome treated non-GSC glioma cells by Notch1 RNA interference and Notch inhibitors. The findings in this study indicated that GSC exosomes act as information carriers, mediated non-GSC glioma cell dedifferentiation into GSCs by delivering Notch1 protein through Notch1 signaling activation, and enhanced stemness and tumorigenicity of non-GSC glioma cells.
Collapse
Affiliation(s)
- Zhen Sun
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Li Wang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Yueling Zhou
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Lihua Dong
- Human Anatomy Department, School of Preclinical and Forensic Medcine, Sichuan University, Chengdu, 610041, China
| | - Weichao Ma
- Neurosurgery Department, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liang Lv
- Neurosurgery Department, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Zhang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Xiujie Wang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China.
| |
Collapse
|
24
|
Wu HB, Huang SS, Lu CG, Tian SD, Chen M. CircAPLP2 regulates the proliferation and metastasis of colorectal cancer by targeting miR-101-3p to activate the Notch signalling pathway. Am J Transl Res 2020; 12:2554-2569. [PMID: 32655790 PMCID: PMC7344090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 04/30/2020] [Indexed: 06/11/2023]
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers, and it has a poor prognosis. Emerging evidence shows that circular RNAs (circRNAs) may act as good therapeutic targets for cancers due to their abundance and stability. However, their regulatory role in CRC needs further investigation. This study revealed that circAPLP2 was upregulated and miR-101-3p was downregulated in CRC tissues and cells compared to normal controls. Knockdown of circAPLP2 and overexpression of miR-101-3p inhibited the cell proliferation, migration and invasion and induced the apoptosis of CRC cells. circAPLP2 acted as a miR-101-3p sponge to upregulate its target gene Notch1, which activated cascades of proliferation- and metastasis-related proteins (c-Myc, cyclin D1, MMP-2 and MMP-9). Additionally, knockdown of circAPLP2 suppressed tumour growth and liver metastases of CRC in nude mice. Taken together, these results indicate that circAPLP2 promotes proliferation and metastasis by targeting miR-101-3p to activate the Notch signalling pathway in CRC, which provides new insights into the mechanisms underlying CRC malignancy and suggests a new therapeutic target.
Collapse
Affiliation(s)
- Han-Bing Wu
- Department of Oncology, The First People’s Hospital of Huaihua CityHuaihua 418000, Hunan, P. R. China
| | - Shi-Si Huang
- Department of Oncology, Central South University Xiangya School of Medicine Affiliated Haikou HospitalHaikou 570208, P. R. China
| | - Chang-Geng Lu
- Department of Oncology, The First People’s Hospital of Huaihua CityHuaihua 418000, Hunan, P. R. China
| | - Shao-Dong Tian
- Department of Oncology, The First People’s Hospital of Huaihua CityHuaihua 418000, Hunan, P. R. China
| | - Ming Chen
- Department of Gastroenterology, The First People’s Hospital of Huaihua CityHuaihua 418000, Hunan, P. R. China
| |
Collapse
|
25
|
Panza S, Russo U, Giordano F, Leggio A, Barone I, Bonofiglio D, Gelsomino L, Malivindi R, Conforti FL, Naimo GD, Giordano C, Catalano S, Andò S. Leptin and Notch Signaling Cooperate in Sustaining Glioblastoma Multiforme Progression. Biomolecules 2020; 10:biom10060886. [PMID: 32526957 PMCID: PMC7356667 DOI: 10.3390/biom10060886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/27/2020] [Accepted: 06/06/2020] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant form of glioma, which represents one of the commonly occurring tumors of the central nervous system. Despite the continuous development of new clinical therapies against this malignancy, it still remains a deadly disease with very poor prognosis. Here, we demonstrated the existence of a biologically active interaction between leptin and Notch signaling pathways that sustains GBM development and progression. We found that the expression of leptin and its receptors was significantly higher in human glioblastoma cells, U-87 MG and T98G, than in a normal human glial cell line, SVG p12, and that activation of leptin signaling induced growth and motility in GBM cells. Interestingly, flow cytometry and real-time RT-PCR assays revealed that GBM cells, grown as neurospheres, displayed stem cell-like properties (CD133+) along with an enhanced expression of leptin receptors. Leptin treatment significantly increased the neurosphere forming efficiency, self-renewal capacity, and mRNA expression levels of the stemness markers CD133, Nestin, SOX2, and GFAP. Mechanistically, we evidenced a leptin-mediated upregulation of Notch 1 receptor and the activation of its downstream effectors and target molecules. Leptin-induced effects on U-87 MG and T98G cells were abrogated by the selective leptin antagonist, the peptide LDFI (Leu-Asp-Phe-Ile), as well as by the specific Notch signaling inhibitor, GSI (Gamma Secretase Inhibitor) and in the presence of a dominant-negative of mastermind-like-1. Overall, these findings demonstrate, for the first time, a functional interaction between leptin and Notch signaling in GBM, highlighting leptin/Notch crosstalk as a potential novel therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Umberto Russo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Antonella Leggio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
- Centro Sanitario, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
- Centro Sanitario, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
- Centro Sanitario, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
- Centro Sanitario, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy
- Correspondence: (S.C.); (S.A.); Tel.: +39-0984-496207 (S.C.); +39-0984-496201 (S.A.)
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy; (S.P.); (U.R.); (F.G.); (A.L.); (I.B.); (D.B.); (L.G.); (R.M.); (F.L.C.); (G.D.N.); (C.G.)
- Centro Sanitario, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende (CS), Italy
- Correspondence: (S.C.); (S.A.); Tel.: +39-0984-496207 (S.C.); +39-0984-496201 (S.A.)
| |
Collapse
|
26
|
Koveitypour Z, Panahi F, Vakilian M, Peymani M, Seyed Forootan F, Nasr Esfahani MH, Ghaedi K. Signaling pathways involved in colorectal cancer progression. Cell Biosci 2019; 9:97. [PMID: 31827763 PMCID: PMC6889432 DOI: 10.1186/s13578-019-0361-4] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the fourth leading cause of the worldwide cancer mortality. Different molecular mechanisms have been attributed to the development and progress of CRC. In this review, we will focus on the mitogen-activated protein kinase (MAPK) cascades downstream of the epidermal growth factor receptor (EGFR), Notch, PI3K/AKT pathway, transforming growth factor-β (TGF-β), and Wnt signaling pathways. Various mutations in the components of these signaling pathways have been linked to the development of CRC. Accordingly, numerous efforts have been carried out to target the signaling pathways to develop novel therapeutic approaches. Herein, we review the signaling pathways involved in the incidence and progression of CRC, and the strategies for the therapy targeting components of signaling pathways in CRC.
Collapse
Affiliation(s)
- Zahra Koveitypour
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran
| | - Farnoush Panahi
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran
| | - Mehrdad Vakilian
- 6Department of Cell Regeneration and Advanced Therapies, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain.,7Department of Cellular Biology, Genetics and Physiology, Faculty of Science, University of Malaga (UMA), Malaga, Spain
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, P.O. Box: 88137-33395, Shahrekord, Iran.,4Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, P.O. Box: 816513-1378, Isfahan, Iran
| | - Farzad Seyed Forootan
- 4Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, P.O. Box: 816513-1378, Isfahan, Iran.,Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Mohammad Hossein Nasr Esfahani
- 4Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, P.O. Box: 816513-1378, Isfahan, Iran
| | - Kamran Ghaedi
- 3Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.,4Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, P.O. Box: 816513-1378, Isfahan, Iran
| |
Collapse
|
27
|
Saurty-Seerunghen MS, Bellenger L, El-Habr EA, Delaunay V, Garnier D, Chneiweiss H, Antoniewski C, Morvan-Dubois G, Junier MP. Capture at the single cell level of metabolic modules distinguishing aggressive and indolent glioblastoma cells. Acta Neuropathol Commun 2019; 7:155. [PMID: 31619292 PMCID: PMC6796454 DOI: 10.1186/s40478-019-0819-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/29/2019] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma cell ability to adapt their functioning to microenvironment changes is a source of the extensive intra-tumor heterogeneity characteristic of this devastating malignant brain tumor. A systemic view of the metabolic pathways underlying glioblastoma cell functioning states is lacking. We analyzed public single cell RNA-sequencing data from glioblastoma surgical resections, which offer the closest available view of tumor cell heterogeneity as encountered at the time of patients’ diagnosis. Unsupervised analyses revealed that information dispersed throughout the cell transcript repertoires encoded the identity of each tumor and masked information related to cell functioning states. Data reduction based on an experimentally-defined signature of transcription factors overcame this hurdle. It allowed cell grouping according to their tumorigenic potential, regardless of their tumor of origin. The approach relevance was validated using independent datasets of glioblastoma cell and tissue transcriptomes, patient-derived cell lines and orthotopic xenografts. Overexpression of genes coding for amino acid and lipid metabolism enzymes involved in anti-oxidative, energetic and cell membrane processes characterized cells with high tumorigenic potential. Modeling of their expression network highlighted the very long chain polyunsaturated fatty acid synthesis pathway at the core of the network. Expression of its most downstream enzymatic component, ELOVL2, was associated with worsened patient survival, and required for cell tumorigenic properties in vivo. Our results demonstrate the power of signature-driven analyses of single cell transcriptomes to obtain an integrated view of metabolic pathways at play within the heterogeneous cell landscape of patient tumors.
Collapse
|
28
|
Mizuguchi A, Yamashita S, Yokogami K, Morishita K, Takeshima H. Ecotropic viral integration site 1 regulates EGFR transcription in glioblastoma cells. J Neurooncol 2019; 145:223-231. [PMID: 31617054 PMCID: PMC6856030 DOI: 10.1007/s11060-019-03310-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Abstract
Purpose Ecotropic viral integration site-1 (EVI1) is a transcription factor that contributes to the unfavorable prognosis of leukemia, some epithelial cancers, and glial tumors. However, the biological function of EVI1 in glioblastoma multiforme (GBM) remains unclear. Based on microarray experiments, EVI1 has been reported to regulate epidermal growth factor receptor (EGFR) transcription. Signal transduction via EGFR plays an essential role in glioblastoma. Therefore, we performed this study to clarify the importance of EVI1 in GBM by focusing on the regulatory mechanism between EVI1 and EGFR transcription. Methods We performed immunohistochemical staining and analyzed the EVI1-expression in glioma tissue. To determine the relationship between EVI1 and EGFR, we induced siRNA-mediated knockdown of EVI1 in GBM cell lines. To investigate the region that was essential for the EVI1 regulation of EGFR expression, we conducted promoter reporter assays. We performed WST-8 assay to investigate whether EVI1 affected on the proliferation of GBM cells or not. Results It was observed that 22% of GBM tissues had over 33% of tumor cells expressing EVI1, whereas no lower-grade glioma tissue had over 33% by immunohistochemistry. In A172 and YKG1 cells, the expression levels of EGFR and EVI1 correlated. Analysis of the EGFR promoter region revealed that the EGFR promoter (from − 377 to − 266 bp) was essential for the EVI regulation of EGFR expression. We showed that EVI1 influenced the proliferation of A172 and YKG1 cells. Conclusion This is the first study reporting the regulation of EGFR transcription by EVI1 in GBM cells. Electronic supplementary material The online version of this article (10.1007/s11060-019-03310-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Asako Mizuguchi
- Department of Neurosurgery, Faculty of Medicine, University of Miyazaki, 5200, Kiyotake-cho, Kihara, Miyazaki-shi, Miyazaki, 889-1601, Japan.
| | - Shinji Yamashita
- Department of Neurosurgery, Faculty of Medicine, University of Miyazaki, 5200, Kiyotake-cho, Kihara, Miyazaki-shi, Miyazaki, 889-1601, Japan
| | - Kiyotaka Yokogami
- Department of Neurosurgery, Faculty of Medicine, University of Miyazaki, 5200, Kiyotake-cho, Kihara, Miyazaki-shi, Miyazaki, 889-1601, Japan
| | - Kazuhiro Morishita
- Department of Tumor and Cellular Biochemistry, Faculty of Medicine, University of Miyazaki, 5200, Kiyotake-cho, Kihara, Miyazaki-shi, Miyazaki, 889-1601, Japan
| | - Hideo Takeshima
- Department of Neurosurgery, Faculty of Medicine, University of Miyazaki, 5200, Kiyotake-cho, Kihara, Miyazaki-shi, Miyazaki, 889-1601, Japan
| |
Collapse
|
29
|
Pozzoli G, Marei HE, Althani A, Boninsegna A, Casalbore P, Marlier LNJL, Lanzilli G, Zonfrillo M, Petrucci G, Rocca B, Navarra P, Sgambato A, Cenciarelli C. Aspirin inhibits cancer stem cells properties and growth of glioblastoma multiforme through Rb1 pathway modulation. J Cell Physiol 2019; 234:15459-15471. [PMID: 30701538 DOI: 10.1002/jcp.28194] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/14/2019] [Indexed: 01/24/2023]
Abstract
Several clinical studies indicated that the daily use of aspirin or acetylsalicylic acid reduces the cancer risk via cyclooxygenases (Cox-1 and Cox-2) inhibition. In addition, aspirin-induced Cox-dependent and -independent antitumor effects have also been described. Here we report, for the first time, that aspirin treatment of human glioblastoma cancer (GBM) stem cells, a small population responsible for tumor progression and recurrence, is associated with reduced cell proliferation and motility. Aspirin did not interfere with cell viability but induced cell-cycle arrest. Exogenous prostaglandin E2 significantly increased cell proliferation but did not abrogate the aspirin-mediated growth inhibition, suggesting a Cox-independent mechanism. These effects appear to be mediated by the increase of p21 waf1 and p27 Kip1 , associated with a reduction of Cyclin D1 and Rb1 protein phosphorylation, and involve the downregulation of key molecules responsible for tumor development, that is, Notch1, Sox2, Stat3, and Survivin. Our results support a possible role of aspirin as adjunctive therapy in the clinical management of GBM patients.
Collapse
Affiliation(s)
- Giacomo Pozzoli
- Institute of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
- Pharmacology Unit, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Alma Boninsegna
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Patrizia Casalbore
- Department of Biomedical Sciences, Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy
| | - Lionel N J L Marlier
- Department of Biomedical Sciences, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Rome, Italy
| | - Giulia Lanzilli
- Department of Biomedical Sciences, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Rome, Italy
| | - Manuela Zonfrillo
- Department of Biomedical Sciences, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Rome, Italy
| | - Giovanna Petrucci
- Institute of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Bianca Rocca
- Institute of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Pierluigi Navarra
- Institute of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
- Pharmacology Unit, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Alessandro Sgambato
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Cenciarelli
- Department of Biomedical Sciences, Institute of Translational Pharmacology (IFT), National Research Council (CNR), Rome, Italy
| |
Collapse
|
30
|
Reliability of intraoperative ultrasound in detecting tumor residual after brain diffuse glioma surgery: a systematic review and meta-analysis. Neurosurg Rev 2019; 43:1221-1233. [PMID: 31410683 DOI: 10.1007/s10143-019-01160-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/28/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Abstract
Intraoperative ultrasonography (iUS) is considered an accurate, safe, and cost-effective tool to estimate the extent of resection of both high-grade (HGG) and low-grade (DLGG) diffuse gliomas (DGs). However, it is currently missing an evidence-based assessment of iUS diagnostic accuracy in DGs surgery. The objective of review is to perform a systematic review and meta-analysis of the diagnostic performance of iUS in detecting tumor residue after DGs resection. A comprehensive literature search for studies published through October 2018 was performed according to PRISMA-DTA and STARD 2015 guidelines, using the following algorithm: ("ultrasound" OR "ultrasonography" OR "ultra-so*" OR "echo*" OR "eco*") AND ("brain" OR "nervous") AND ("tumor" OR "tumour" OR "lesion" OR "mass" OR "glio*" OR "GBM") AND ("surgery" OR "surgical" OR "microsurg*" OR "neurosurg*"). Pooled sensitivity, specificity, positive and negative likelihood ratios (LR+ and LR-), and diagnostic odds ratio (DOR) of iUS in DGs were calculated. A subgroup analysis for HGGs and DLGGs was also conducted. Thirteen studies were included in the systematic review (665 DGs). Ten articles (409 DGs) were selected for the meta-analysis with the following results: sensitivity 72.2%, specificity 93.5%, LR- 0.29, LR+ 3, and DOR 9.67. Heterogeneity among studies was non-significant. Subgroup analysis demonstrates a better diagnostic performance of iUS for DLGGs compared with HGGs. iUS is an effective technique in assessing DGs resection. No significant differences are seen regarding iUS modality and transducer characteristics. Its diagnostic performance is higher in DLGGs than HGGs and could be worsened by previous treatments, surgical artifacts, and small tumor residue volumes.
Collapse
|
31
|
Yi L, Zhou X, Li T, Liu P, Hai L, Tong L, Ma H, Tao Z, Xie Y, Zhang C, Yu S, Yang X. Notch1 signaling pathway promotes invasion, self-renewal and growth of glioma initiating cells via modulating chemokine system CXCL12/CXCR4. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:339. [PMID: 31382985 PMCID: PMC6683584 DOI: 10.1186/s13046-019-1319-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/10/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Glioma initiating cells (GICs), also known as glioma stem cells (GSCs), play an important role in the progression and recurrence of glioblastoma multiforme (GBM) due to their potential for self-renewal, multiple differentiation and tumor initiation. In the recent years, Notch1 has been found to be overexpressed in GICs. However, the regulatory mechanism of Notch1 in the self-renewal and invasion ability of GICs remains unclear. This study aims to explore the effect of Notch pathway on self-renewal and invasion of GICs and the underlying mechanisms. METHODS Bioinformatic analysis and immunohistochemistry (IHC) were performed to evaluate the expression of Notch1 and Hes1 in GBM samples. Immunofluorescent (IF) staining was performed to observe the distribution of Notch1 and CXCR4 in GBM and GICs. Both pharmacological intervention and RNA interference were employed to investigate the role of Notch1 in GICs self-renewal, invasion and tumor growth in vitro or in vivo. The crosstalk effect of Notch1 and CXCL12/CXCR4 system on GIC self-renewal and invasion was explored by sphere formation assay, limiting dilution assay and Transwell assay. Western blots were used to verify the activation of Notch1/CXCR4/AKT pathway in self-renewal, invasion and tumor growth of GICs. Luciferase reporter assay was used to testify the potential binding site of Notch1 signaling and CXCR4. The orthotopic GICs implantations were established to analyze the role and the mechanism of Notch1 in glioma progression in vivo. RESULTS Notch1 signaling activity was elevated in GBM tissues. Notch1 and CXCR4 were both upregulated in GICs, compared to Notch1 positive glioma cells comprised a large proportion in the CD133+ glioma cell spheres, CXCR4 positive glioma cells which usually expressed Notch1 both and dispersed in the periphery of the sphere, only represent a small subset of CD133+ glioma cell spheres. Furthermore, downregulation of the Notch1 pathway by shRNA and MK0752 significantly inhibited the PI3K/AKT/mTOR signaling pathway via the decreased expression of CXCR4 in GICs, and weakened the self-renewal, invasion and tumor growth ability of GICs. CONCLUSIONS These findings suggest that the cross-talk between Notch1 signaling and CXCL12/CXCR4 system could contribute to the self-renewal and invasion of GICs, and this discovery could help drive the design of more effective therapies in Notch1-targeted treatment of GBMs.
Collapse
Affiliation(s)
- Li Yi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Xingchen Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Bengbu Medical College, Anhui, 233000, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Peidong Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Long Hai
- Department of Radiation Oncology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Henan, 450000, China
| | - Luqing Tong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Haiwen Ma
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Zhennan Tao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Yang Xie
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Chen Zhang
- Neuro-Oncology Department, The University of Texas MD Anderson Cancer Center, Houston, 77030, Texas, USA
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China. .,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.
| |
Collapse
|
32
|
Yu J, Canalis E. The Hajdu Cheney mutation sensitizes mice to the osteolytic actions of tumor necrosis factor α. J Biol Chem 2019; 294:14203-14214. [PMID: 31371452 DOI: 10.1074/jbc.ra119.009824] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/29/2019] [Indexed: 11/06/2022] Open
Abstract
Hajdu Cheney syndrome (HCS) is characterized by craniofacial developmental abnormalities, acro-osteolysis, and osteoporosis and is associated with gain-of-NOTCH2 function mutations. A mouse model of HCS termed Notch2tm1.1Ecan harboring a mutation in exon 34 of Notch2 replicating the one found in HCS was used to determine whether the HCS mutation sensitizes the skeleton to the osteolytic effects of tumor necrosis factor α (TNFα). TNFα injected over the calvarial vault caused a greater increase in osteoclast number, osteoclast surface, and eroded surface in Notch2tm1.1Ecan mice compared with littermate WT controls. Accordingly, the effect of TNFα on osteoclastogenesis was greatly enhanced in cultures of bone marrow-derived macrophages (BMMs) from Notch2tm1.1Ecan mice when compared with the activity of TNFα in control cultures. TNFα induced the expression of Notch2 and Notch2 mutant mRNA by ∼2-fold, possibly amplifying the NOTCH2-dependent induction of osteoclastogenesis. The effect of TNFα on osteoclastogenesis in Notch2tm1.1Ecan mutants depended on NOTCH2 activation because it was reversed by anti-NOTCH2 negative regulatory region and anti-jagged 1 antibodies. The inactivation of Hes1 prevented the TNFα effect on osteoclastogenesis in the context of the Notch2tm1.1Ecan mutation. In addition, the induction of Il1b, but not of Tnfa and Il6, mRNA by TNFα was greater in Notch2tm1.1Ecan BMMs than in control cells, possibly contributing to the actions of TNFα and NOTCH2 on osteoclastogenesis. In conclusion, the HCS mutation enhances TNFα-induced osteoclastogenesis and the inflammatory bone-resorptive response possibly explaining the acro-osteolysis observed in affected individuals.
Collapse
Affiliation(s)
- Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut 06030.,UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| | - Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut 06030 .,UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030.,Department of Medicine, UConn Health, Farmington, Connecticut 06030
| |
Collapse
|
33
|
Hoarau-Véchot J, Touboul C, Halabi N, Blot-Dupin M, Lis R, Abi Khalil C, Rafii S, Rafii A, Pasquier J. Akt-activated endothelium promotes ovarian cancer proliferation through notch activation. J Transl Med 2019; 17:194. [PMID: 31182109 PMCID: PMC6558713 DOI: 10.1186/s12967-019-1942-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/30/2019] [Indexed: 12/17/2022] Open
Abstract
Background One main challenge in ovarian cancer rests on the presence of a relapse and an important metastatic disease, despite extensive surgical debulking and chemotherapy. The difficulty in containing metastatic cancer is partly due to the heterotypic interaction of tumor and its microenvironment. In this context, evidence suggests that endothelial cells (EC) play an important role in ovarian tumor growth and chemoresistance. Here, we studied the role of tumor endothelium on ovarian cancer cells (OCCs). Methods We evaluated the effect of activated endothelial cells on ovarian cancer cell proliferation and resistance to chemotherapy and investigated the survival pathways activated by endothelial co-culture. Results The co-culture between OCCs and E4+ECs, induced an increase of OCCs proliferation both in vitro and in vivo. This co-culture induced an increase of Notch receptors expression on OCC surface and an increase of Jagged 1 expression on E4+ECs surface and activation of survival pathways leading to chemoresistance by E4+ECs. Conclusion The targeting of aberrant NOTCH signaling could constitute a strategy to disrupt the pro-tumoral endothelial niche.
Collapse
Affiliation(s)
- Jessica Hoarau-Véchot
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar.,Epigenetics Cardiovascular Laboratory, Department of Genetic Medicine, Weill Cornell Medicine-Qatar, PO box 24144, Doha, Qatar
| | - Cyril Touboul
- INSERM U955, Equipe 7, Créteil, France.,Faculté de Médecine de Créteil UPEC-Paris XII, Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000, Créteil, France
| | - Najeeb Halabi
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar.,Department of Genetic Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Morgane Blot-Dupin
- Faculté de Médecine de Créteil UPEC-Paris XII, Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000, Créteil, France
| | - Raphael Lis
- Department of Genetic Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Charbel Abi Khalil
- Epigenetics Cardiovascular Laboratory, Department of Genetic Medicine, Weill Cornell Medicine-Qatar, PO box 24144, Doha, Qatar.,Department of Genetic Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Shahin Rafii
- Department of Genetic Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Arash Rafii
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar. .,Department of Genetic Medicine, Weill Cornell Medicine, New York City, NY, USA. .,Department of Gynecologic Oncology, Hospital Foch, Surresnes, France. .,Department of Genetic Medicine and Obstetrics and Gynecology, Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Qatar-Foundation, PO: 24144, Doha, Qatar.
| | - Jennifer Pasquier
- Stem Cell and Microenvironment Laboratory, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar. .,INSERM U955, Equipe 7, Créteil, France.
| |
Collapse
|
34
|
Mori M, Mori T, Yamamoto A, Takagi S, Ueda M. Proliferation of poorly differentiated endometrial cancer cells through autocrine activation of FGF receptor and HES1 expression. Hum Cell 2019; 32:367-378. [PMID: 30963412 DOI: 10.1007/s13577-019-00249-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
Patients with poorly differentiated endometrial cancer show poor prognosis, and effective molecular target-based therapies are needed. Endometrial cancer cells proliferate depending on the activation of HES1 (hairy and enhancer of split-1), which is induced by several pathways, such as the Notch and fibroblast growth factor receptor (FGFR) signaling pathways. In addition, aberrant, ligand-free activation of the FGFR signaling pathway resulting from mutations in FGFR2 was also reported in endometrial cancer. However, a clinical trial showed that there was no difference in the effectiveness of FGFR inhibitors between patients with and without the FGFR2 mutation, suggesting a presence of another signaling pathway for the FGFR activation. Here, we investigated the signaling pathway regulating the expression of HES1 and proliferation of poorly and well-differentiated endometrial cancer cell lines Ishikawa and HEC-50B, respectively. Whereas Ishikawa cells proliferated and expressed HES1 in a Notch signaling-dependent manner, Notch signaling was not involved in HES1 and proliferation of HEC-50B cells. The FGFR inhibitor, NVP-BGJ398, decreased HES1 expression and proliferation of HEC-50B cells; however, HEC50B cells had no mutations in the FGFR2 gene. Instead, HEC-50B cells highly expressed ligands for FGFR2, suggesting that FGFR2 is activated by an autocrine manner, not by ligand-free activation. This autocrine pathway activated Akt downstream of FGFR for cell proliferation. Our findings suggest the usefulness of HES1 as a marker for the proliferation signaling and that FGFR inhibitor may be effective for poorly differentiated endometrial cancers that harbor wild-type FGFR.
Collapse
Affiliation(s)
- Michihiro Mori
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan. .,Kake Institute of Cytopathology, Okayama, Japan.
| | - Toshinori Mori
- Department of Clinical Laboratory, Mihara Medical Associations Hospital, Hiroshima, Japan.,Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Aina Yamamoto
- Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Shoji Takagi
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan.,Kake Institute of Cytopathology, Okayama, Japan
| | - Masatsugu Ueda
- Faculty of Health Sciences, Kio University, Nara, Japan.,Graduate School of Health Sciences, Kio University, Nara, Japan
| |
Collapse
|
35
|
Tao Z, Li T, Ma H, Yang Y, Zhang C, Hai L, Liu P, Yuan F, Li J, Yi L, Tong L, Wang Y, Xie Y, Ming H, Yu S, Yang X. Autophagy suppresses self-renewal ability and tumorigenicity of glioma-initiating cells and promotes Notch1 degradation. Cell Death Dis 2018; 9:1063. [PMID: 30337536 PMCID: PMC6194143 DOI: 10.1038/s41419-018-0957-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 06/01/2018] [Accepted: 06/06/2018] [Indexed: 01/05/2023]
Abstract
Autophagy is a vital process that involves degradation of long-lived proteins and dysfunctional organelles and contributes to cellular metabolism. Glioma-initiating cells (GICs) have the ability to self-renew, differentiate into heterogeneous types of tumor cells, and sustain tumorigenicity; thus, GICs lead to tumor recurrence. Accumulating evidence indicates that autophagy can induce stem cell differentiation and increase the lethality of temozolomide against GICs. However, the mechanism underlying the regulation of GIC self-renewal by autophagy remains uncharacterized. In the present study, autophagy induced by AZD8055 and rapamycin treatment suppressed GIC self-renewal in vitro. We found that autophagy inhibited Notch1 pathway activation. Moreover, autophagy activated Notch1 degradation, which is associated with maintenance of the self-renewal ability of GICs. Furthermore, autophagy abolished the tumorigenicity of CD133 + U87-MG neurosphere cells in an intracranial model. These findings suggest that autophagy regulating GICs self-renewal and tumorigenicity is probably bound up with Notch1 degradation. The results of this study could aid in the design of autophagy-based clinical trials for glioma treatments, which may be of great value.
Collapse
Affiliation(s)
- Zhennan Tao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Haiwen Ma
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Yihan Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Chen Zhang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Long Hai
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Peidong Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Feng Yuan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Jiabo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Li Yi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Luqing Tong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China.,Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Yingshuai Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yang Xie
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Haolang Ming
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300052, China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
36
|
Lu H, Jiang J, Gao Y. The cloning and activity of human Hes1 gene promoter. Mol Med Rep 2017; 17:3164-3169. [PMID: 29257279 DOI: 10.3892/mmr.2017.8240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/15/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the current study was to obtain and analyze the activity of the human Hes1 gene promoter. The genomic DNA of human HeLa cell was used as template, polymerase chain reaction (PCR) was used to amplify the 5' end sequence of Hes1 gene and then the amplified segment was connected to pMD18‑T vector. Subsequently, double enzyme digestion was used for identification and the sequence was detected; the promoter with the correct sequence was inserted into pGL3‑Basic, and the sequence was identified by double enzyme digestion. The recombinant DNA with correct sequence was transiently transfected into cervical cancer cells, and the dual luciferase reporter gene assay system was used to detect the activity of the promoter. The results demonstrated that the human Hes1 gene promoter amplified by PCR was the same as that of the sequence in the gene bank, and the dual luciferase reporter gene assay system demonstrated that there was promoter activity in cervical cancer cells. In conclusion, the Hes1 luciferase reporter recombinant vector was successfully established and transfected into HeLa cells to verify that it has promoter activity, and the core area of the promoter has several tumor‑promoting and tumor suppressor genes. This provides a basis for understanding the regulatory mechanism of Hes1 transcription and translation.
Collapse
Affiliation(s)
- Hai Lu
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Jinqun Jiang
- Clinical Laboratory, Yuebei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
37
|
Li L, Li Y, Wang L, Wu Z, Ma H, Shao J, Li D, Yu H, Nian W, Wang D. Inhibition of Hes1 enhances lapatinib sensitivity in gastric cancer sphere-forming cells. Oncol Lett 2017; 14:3989-3996. [PMID: 28959362 PMCID: PMC5607651 DOI: 10.3892/ol.2017.6683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 05/26/2017] [Indexed: 01/26/2023] Open
Abstract
It has been considered that the neurogenic locus notch homolog protein (Notch) signaling pathway serves an essential role in cellular differentiation, proliferation and apoptosis. However, the function of the Notch signaling pathway in gastric cancer stem cells (GCSCs) and epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) sensitivity remains unclear. The present study aimed to delineate the role of the Notch1 signaling pathway in GCSCs and lapatinib sensitivity. Sphere-forming cells were separated from human gastric cancer MKN45 parental cells. The sphere-forming cells exhibited characteristics of CSCs and higher Notch1 expression compared with that of parental cells. To investigate the role of the Notch1 signaling pathway in GCSCs, the expression of transcription factor Hes1 (Hes1) was knocked down using small interfering RNA against Hes1. It was observed that Hes1 expression was significantly downregulated in knocked down cells. The inhibition of Hes1 suppressed the properties of CSCs, as indicated by significant decreases in the expression of the transcription factor sex determining region Y-box 2, epithelial cell adhesion molecule and the homeobox protein Nanog and reduced spheroid colony formation. In addition, epithelial-mesenchymal transition was significantly impaired in sphere-forming cells following Hes1 knockdown. Furthermore, the inhibition of Hes1 effectively enhanced lapatinib sensitivity in sphere-forming cells. These results suggest that sphere-forming gastric cancer cells possess the characteristics of CSCs, and that the Notch1 signaling pathway serves an essential role in the maintenance of CSCs and lapatinib sensitivity.
Collapse
Affiliation(s)
- Luchun Li
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Yan Li
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Lulu Wang
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Zhijuan Wu
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Huiwen Ma
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Jianghe Shao
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Dairong Li
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Huiqing Yu
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Weiqi Nian
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| | - Donglin Wang
- Department of Oncology, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, P.R. China
| |
Collapse
|