1
|
Lisek M, Tomczak J, Swiatek J, Kaluza A, Boczek T. Histone Deacetylases in Retinoblastoma. Int J Mol Sci 2024; 25:6910. [PMID: 39000021 PMCID: PMC11241206 DOI: 10.3390/ijms25136910] [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: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Retinoblastoma, a pediatric ocular malignancy, presents significant challenges in comprehending its molecular underpinnings and targeted therapeutic approaches. The dysregulated activity of histone deacetylases (HDACs) has been associated with retinoblastoma pathogenesis, influencing critical cellular processes like cell cycle regulation or retinal ganglion cell apoptosis. Through their deacetylase activity, HDACs exert control over key tumor suppressors and oncogenes, influencing the delicate equilibrium between proliferation and cell death. Furthermore, the interplay between HDACs and the retinoblastoma protein pathway, a pivotal aspect of retinoblastoma etiology, reveals a complex network of interactions influencing the tumor microenvironment. The examination of HDAC inhibitors, encompassing both established and novel compounds, offers insights into potential approaches to restore acetylation balance and impede retinoblastoma progression. Moreover, the identification of specific HDAC isoforms exhibiting varying expression in retinoblastoma provides avenues for personalized therapeutic strategies, allowing for interventions tailored to individual patient profiles. This review focuses on the intricate interrelationship between HDACs and retinoblastoma, shedding light on epigenetic mechanisms that control tumor development and progression. The exploration of HDAC-targeted therapies underscores the potential for innovative treatment modalities in the pursuit of more efficacious and personalized management strategies for this disease.
Collapse
Affiliation(s)
- Malwina Lisek
- Department of Molecular Neurochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (J.T.); (J.S.); (A.K.)
| | | | | | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (J.T.); (J.S.); (A.K.)
| |
Collapse
|
2
|
Balaji S, Rao A, Saraswathi KK, Sethu Nagarajan R, Santhi R, Kim U, Muthukkaruppan V, Vanniarajan A. Focused cancer pathway analysis revealed unique therapeutic targets in retinoblastoma. Med Oncol 2024; 41:168. [PMID: 38834895 DOI: 10.1007/s12032-024-02391-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024]
Abstract
Retinoblastoma (RB) is a pediatric cancer of the eye that occurs in 1/15000 live births worldwide. Albeit RB is initiated by the inactivation of RB1 gene, the disease progression relies largely on transcriptional alterations. Therefore, evaluating gene expression is vital to unveil the therapeutic targets in RB management. In this study, we employed an RT2 Profiler™ PCR array for a focused analysis of 84 cancer-specific genes in RB. An interaction network was built with gene expression data to identify the dysregulated pathways in RB. The key transcript alterations identified in 13 tumors by RT2 Profiler™ PCR array was further validated in 15 tumors by independent RT-qPCR. Out of 84 cancer-specific genes, 68 were dysregulated in RB tumors. Among the 68 genes, 23 were chosen for further analysis based on statistical significance and abundance across multiple tumors. Pathway analysis of altered genes showed the frequent perturbations of cell cycle, angiogenesis and apoptotic pathways in RB. Notably, upregulation of MCM2, MKI67, PGF, WEE1, CDC20 and downregulation of COX5A were found in all the tumors. Western blot confirmed the dysregulation of identified targets at protein levels as well. These alterations were more prominent in invasive RB, correlating with the disease pathogenesis. Our molecular analysis thus identified the potential therapeutic targets for improving retinoblastoma treatment. We also suggest that PCR array can be used as a tool for rapid and cost-effective gene expression analysis.
Collapse
Affiliation(s)
- Sekaran Balaji
- Department of Molecular Genetics, Aravind Medical Research Foundation, 1, Anna Nagar, Madurai, Tamil Nadu, 625 020, India
| | - Anindita Rao
- Department of Molecular Genetics, Aravind Medical Research Foundation, 1, Anna Nagar, Madurai, Tamil Nadu, 625 020, India
| | - Karuvel Kannan Saraswathi
- Department of Molecular Genetics, Aravind Medical Research Foundation, 1, Anna Nagar, Madurai, Tamil Nadu, 625 020, India
- Department of Molecular Biology, Aravind Medical Research Foundation - Affiliated to Alagappa University, Karaikudi, Tamil Nadu, 630003, India
| | - Rathinavel Sethu Nagarajan
- Department of Molecular Genetics, Aravind Medical Research Foundation, 1, Anna Nagar, Madurai, Tamil Nadu, 625 020, India
- Department of Molecular Biology, Aravind Medical Research Foundation - Affiliated to Alagappa University, Karaikudi, Tamil Nadu, 630003, India
| | - Radhakrishnan Santhi
- Department of Pathology, Aravind Eye Hospital, Madurai, Tamil Nadu, 625 020, India
| | - Usha Kim
- Department of Orbit, Oculoplasty and Ocular Oncology, Aravind Eye Hospital, Madurai, Tamil Nadu, 625 020, India
| | - Veerappan Muthukkaruppan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625 020, India
| | - Ayyasamy Vanniarajan
- Department of Molecular Genetics, Aravind Medical Research Foundation, 1, Anna Nagar, Madurai, Tamil Nadu, 625 020, India.
- Department of Molecular Biology, Aravind Medical Research Foundation - Affiliated to Alagappa University, Karaikudi, Tamil Nadu, 630003, India.
| |
Collapse
|
3
|
Cobrinik D. Retinoblastoma Origins and Destinations. N Engl J Med 2024; 390:1408-1419. [PMID: 38631004 DOI: 10.1056/nejmra1803083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Affiliation(s)
- David Cobrinik
- From the Vision Center, Department of Surgery, and Saban Research Institute, Children's Hospital Los Angeles, and the Departments of Ophthalmology and Biochemistry and Molecular Medicine, Roski Eye Institute, and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California - both in Los Angeles
| |
Collapse
|
4
|
Al-Ghazzawi K, Wessolly M, Dalbah S, Ketteler P, Kiefer T, Bechrakis N, Leyla J, Ting S, Biewald E, Mairinger FD. PDGF, NGF, and EGF as main contributors to tumorigenesis in high-risk retinoblastoma. Front Oncol 2023; 13:1144951. [PMID: 37965463 PMCID: PMC10642836 DOI: 10.3389/fonc.2023.1144951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 09/12/2023] [Indexed: 11/16/2023] Open
Abstract
Retinoblastoma (RB) is the most common form of eye cancer experienced in childhood. Its aggressive malignancy is associated with excellent survival rates in high-income countries; however, the prognosis in third-world countries is less favorable. Early diagnosis can maximize the patient's visual outcomes and their survival rate. Therapy should be conducted in highly specialized treatment centers. Intravenous chemotherapy (IVC) in bilaterally affected children currently forms the majority of therapy. Local destructive procedures and local chemotherapies such as intra-arterial chemotherapy (IAC) or intravitreal chemotherapy can be taken into consideration depending on the extent and size of the tumor. Nonetheless, children and parents remain under constant stress, revisiting doctors for medical treatment and fearing vision loss and even enucleation of the eye. Adequate molecular patient stratification to improve targeted therapy is still lacking. This retrospective study analyzed formalin-fixed paraffin-embedded specimens from a cohort of 21 RB samples. A total of 11 of those samples showed undifferentiated retinoblastoma (URB) histopathological risk features, and the other 10 showed differentiated retinoblastoma (DRB) histopathological grading. RNA from all samples was isolated and analyzed via digital gene expression patterns. Conductors of cell survival and DNA repair were dominant in the DRB samples. In contrast, the agents responsible for cell-cycle progression and apoptosis were overexpressed in URB samples. Our work reveals the importance of molecular mechanisms within the immune system subjected to histologic subtypes of RB, providing more detailed background on their genetic behavior. This is of great interest for therapeutic strategies, such as targeted immune- and gene-based therapies, for retinoblastoma.
Collapse
Affiliation(s)
- Karim Al-Ghazzawi
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - Michael Wessolly
- Department of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sami Dalbah
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - Petra Ketteler
- Department of Pediatric Hematology and Oncology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tobias Kiefer
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | | | - Jabbarli Leyla
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - Saskia Ting
- Institute of Pathology Nordhessen, Kassel, Germany
| | - Eva Biewald
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - Fabian D. Mairinger
- Department of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
5
|
Norrie JL, Lupo M, Shirinifard A, Djekidel N, Ramirez C, Xu B, Dundee JM, Dyer MA. Latent Epigenetic Programs in Müller Glia Contribute to Stress, Injury, and Disease Response in the Retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.15.562396. [PMID: 37905050 PMCID: PMC10614790 DOI: 10.1101/2023.10.15.562396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Previous studies have demonstrated the dynamic changes in chromatin structure during retinal development that correlate with changes in gene expression. However, a major limitation of those prior studies was the lack of cellular resolution. Here, we integrate single-cell (sc) RNA-seq and scATAC-seq with bulk retinal data sets to identify cell type-specific changes in the chromatin structure during development. Although most genes' promoter activity is strongly correlated with chromatin accessibility, we discovered several hundred genes that were transcriptionally silent but had accessible chromatin at their promoters. Most of those silent/accessible gene promoters were in the Müller glial cells. The Müller cells are radial glia of the retina and perform a variety of essential functions to maintain retinal homeostasis and respond to stress, injury, or disease. The silent/accessible genes in Müller glia are enriched in pathways related to inflammation, angiogenesis, and other types of cell-cell signaling and were rapidly activated when we tested 15 different physiologically relevant conditions to mimic retinal stress, injury, or disease in human and murine retinae. We refer to these as "pliancy genes" because they allow the Müller glia to rapidly change their gene expression and cellular state in response to different types of retinal insults. The Müller glial cell pliancy program is established during development, and we demonstrate that pliancy genes are necessary and sufficient for regulating inflammation in the murine retina in vivo. In zebrafish, Müller glia can de-differentiate and form retinal progenitor cells that replace lost neurons. The pro-inflammatory pliancy gene cascade is not activated in zebrafish Müller glia following injury, and we propose a model in which species-specific pliancy programs underly the differential response to retinal damage in species that can regenerate retinal neurons (zebrafish) versus those that cannot (humans and mice).
Collapse
|
6
|
Cheng YM, Ma C, Jin K, Jin ZB. Retinal organoid and gene editing for basic and translational research. Vision Res 2023; 210:108273. [PMID: 37307693 DOI: 10.1016/j.visres.2023.108273] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023]
Abstract
The rapid evolution of two technologies has greatly transformed the basic, translational, and clinical research in the mammalian retina. One is the retinal organoid (RO) technology. Various induction methods have been created or adapted to generate species-specific, disease-specific, and experimental-targeted retinal organoids (ROs). The process of generating ROs can highly mimic the in vivo retinal development, and consequently, the ROs resemble the retina in many aspects including the molecular and cellular profiles. The other technology is the gene editing, represented by the classical CRISPR-Cas9 editing and its derivatives such as prime editing, homology independent targeted integration (HITI), base editing and others. The combination of ROs and gene editing has opened up countless possibilities in the study of retinal development, pathogenesis, and therapeutics. We review recent advances in the ROs, gene editing methodologies, delivery vectors, and related topics that are particularly relevant to retinal studies.
Collapse
Affiliation(s)
- You-Min Cheng
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730 China
| | - Chao Ma
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730 China
| | - Kangxin Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730 China.
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730 China.
| |
Collapse
|
7
|
Zhao H, Du C, Yang G, Wang Y. Diagnosis, treatment, and research status of rare diseases related to birth defects. Intractable Rare Dis Res 2023; 12:148-160. [PMID: 37662624 PMCID: PMC10468410 DOI: 10.5582/irdr.2023.01052] [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: 06/29/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023] Open
Abstract
Rare diseases are diseases that occur at low prevalence, and most of them are chronic and serious diseases that are often life-threatening. Currently, there is no unified definition for rare diseases. The diagnosis, treatment, and research of rare diseases have become the focus of medicine and biopharmacology, as well as the breakthrough point of clinical and basic research. Birth defects are the hard-hit area of rare diseases and the frontiers of its research. Since most of these defects have a genetic basis, early screening and diagnosis have important scientific value and social significance for the prevention and control of such diseases. At present, there is no effective treatment for most rare diseases, but progress in prenatal diagnosis and screening can prevent the occurrence of diseases and help prevent and treat rare diseases. This article discusses the progress in genetic-related birth defects and rare diseases.
Collapse
Affiliation(s)
- Hongjuan Zhao
- Department of Gynecology and Obstetrics, Shandong Provincial Third Hospital, Shandong University, Ji'nan, China
| | - Chen Du
- Department of Gynecology and Obstetrics, Inner Mongolia Medical University Affiliated Hospital, Hohhot, China
| | - Guang Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Gynecology and Obstetrics, Inner Mongolia Medical University Affiliated Hospital, Hohhot, China
| |
Collapse
|
8
|
Retinoblastoma: From genes to patient care. Eur J Med Genet 2022; 66:104674. [PMID: 36470558 DOI: 10.1016/j.ejmg.2022.104674] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/04/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Retinoblastoma is the most common paediatric neoplasm of the retina, and one of the earliest model of cancer genetics since the identification of the master tumour suppressor gene RB1. Tumorigenesis has been shown to be driven by pathogenic variants of the RB1 locus, but also genomic and epigenomic alterations outside the locus. The increasing knowledge on this "mutational landscape" is used in current practice for precise genetic testing and counselling. Novel methods provide access to pre-therapeutic tumour DNA, by isolating cell-free DNA from aqueous humour or plasma. This is expected to facilitate assessment of the constitutional status of RB1, to provide an early risk stratification using molecular prognostic markers, to follow the response to the treatment in longitudinal studies, and to predict the response to targeted therapies. The aim of this review is to show how molecular genetics of retinoblastoma drives diagnosis, treatment, monitoring of the disease and surveillance of the patients and relatives. We first recap the current knowledge on retinoblastoma genetics and its use in every-day practice. We then focus on retinoblastoma subgrouping at the era of molecular biology, and the expected input of cell-free DNA in the field.
Collapse
|
9
|
Zhao H, Collet C, Peng D, Sinha UK, Lin DC. Investigation of early neoplastic transformation and premalignant biology using genetically engineered organoid models. Comput Struct Biotechnol J 2022; 20:5309-5315. [PMID: 36212534 PMCID: PMC9513696 DOI: 10.1016/j.csbj.2022.09.026] [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: 07/08/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022] Open
Abstract
Organoid modeling is a powerful, robust and efficient technology faithfully preserving physiological and pathological characteristics of tissues of origin. Recently, substantial advances have been made in applying genetically engineered organoid models to study early tumorigenesis and premalignant biology. These efforts promise to identify novel avenues for early cancer detection, intervention and prevention. Here, we highlight significant advancements in the functional characterization of early genomic and epigenomic events during neoplastic evolution using organoid modeling, discuss the application of the lineage-tracing methodology in organoids to study cancer cells-of-origin, and review future opportunities for further development and improvement of organoid modeling of cancer precursors.
Collapse
Affiliation(s)
- Hua Zhao
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Casey Collet
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Dongzi Peng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Uttam K. Sinha
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
10
|
Nam C, Ziman B, Sheth M, Zhao H, Lin DC. Genomic and Epigenomic Characterization of Tumor Organoid Models. Cancers (Basel) 2022; 14:4090. [PMID: 36077628 PMCID: PMC9454968 DOI: 10.3390/cancers14174090] [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: 08/03/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Tumor organoid modeling has been recognized as a state-of-the-art system for in vitro research on cancer biology and precision oncology. Organoid culture technologies offer distinctive advantages, including faithful maintenance of physiological and pathological characteristics of human disease, self-organization into three-dimensional multicellular structures, and preservation of genomic and epigenomic landscapes of the originating tumor. These features effectively position organoid modeling between traditional cell line cultures in two dimensions and in vivo animal models as a valid, versatile, and robust system for cancer research. Here, we review recent advances in genomic and epigenomic characterization of tumor organoids and the novel findings obtained, highlight significant progressions achieved in organoid modeling of gene-drug interactions and genotype-phenotype associations, and offer perspectives on future opportunities for organoid modeling in basic and clinical cancer research.
Collapse
Affiliation(s)
| | | | | | | | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
11
|
Xu N, Cui Y, Shi H, Guo G, Sun F, Jian T, Rao H. UBE2T/STAT3 Signaling Promotes the Proliferation and Tumorigenesis in Retinoblastoma. Invest Ophthalmol Vis Sci 2022; 63:20. [PMID: 35980647 PMCID: PMC9404369 DOI: 10.1167/iovs.63.9.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this paper was to investigate the expression and function of Ubiquitin-conjugating enzyme 2T (UBE2T), a human E2 ubiquitin-conjugating enzyme, in human retinoblastoma. Methods The expression of UBE2T in normal retina and retinoblastoma was analyzed using the Gene Expression Omnibus (GEO) databases, and its expression was immunohistochemically evaluated in 29 retinoblastoma sections and 5 normal retinas. Then CCK-8, flow cytometry, RNA-sequencing analysis, and in vivo assays were performed to explore the exact role of UBE2T in retinoblastoma. Results We found that retinoblastoma showed higher UBE2T expression than normal retina in GEO datasets and tissues. The immunoreactive score of UBE2T ≥4 was associated with group E in IIRC, T2-T4b in pTNM staging, poorly differentiated retinoblastoma, and high-risk histopathological factors. Knockdown of UBE2T reduced the cell viability, increased the apoptosis cells and G0/G1 cells, and inhibited subcutaneous tumor growth in vivo. Mechanistic studies showed that UBE2T knockdown induced down-regulation of phosphorylation of STAT3 and its downstream genes in vitro and in vivo. Rescue assays confirmed that STAT3 signaling pathway was involved in the effect of reduced cell viability, elevated apoptosis cells, and G0/G1 cells mediated by UBE2T knockdown. Conclusions Our data indicate that UBE2T significantly participates in the proliferation of retinoblastoma via the STAT3 signaling pathway, suggesting the potential of UBE2T as a therapeutic target for retinoblastoma treatment.
Collapse
Affiliation(s)
- Nuo Xu
- Department of Ophthalmology, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China.,Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.,Department of Oculoplastic and Orbital Diseases, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Yi Cui
- Department of Ophthalmology, Fujian Medical University Union Hospital, Tianjin, China
| | - Hong Shi
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Guodong Guo
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Fengyuan Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.,Department of Oculoplastic and Orbital Diseases, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Tianming Jian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.,Department of Oculoplastic and Orbital Diseases, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Huiying Rao
- Department of Ophthalmology, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
12
|
Azimi F, Mirshahi R, Naseripour M. Review: New horizons in retinoblastoma treatment: an updated review article. Mol Vis 2022; 28:130-146. [PMID: 36034735 PMCID: PMC9352364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 07/09/2022] [Indexed: 10/25/2022] Open
Abstract
Retinoblastoma (Rb) is a rare childhood intraocular malignancy with an incidence rate of approximately 9000 children per year worldwide. The management of Rb is inherently complex and depends on several factors. The orders of priorities in the treatment of Rb are saving life, globe salvage and vision salvage. Rarity and the young age at diagnosis impede conducting randomized clinical trials (RCTs) for new therapeutic options, and therefore pre-RCTs studies are needed. This review provides an overview of advances in Rb treatment options, focusing on the emergence of new small molecules to treat Rb. Articles related to the management and treatments of Rb were searched in different databases. Several studies and animal models discussing recent advances in the treatment of Rb were included to have a better grasp of the biological mechanisms of Rb. Over the years, the principles of management and treatment of Rb have changed significantly. Innovations in targeted therapies and molecular biology have led to improved patient and ocular survival. However, there is still a need for further evaluation of the long-term effects of these new treatments.
Collapse
Affiliation(s)
- Fatemeh Azimi
- Eye Research Center, the Five Senses Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Mirshahi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masood Naseripour
- Eye Research Center, the Five Senses Institute, Iran University of Medical Sciences, Tehran, Iran,Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Single-cell characterization of malignant phenotypes and microenvironment alteration in retinoblastoma. Cell Death Dis 2022; 13:438. [PMID: 35523772 PMCID: PMC9076657 DOI: 10.1038/s41419-022-04904-8] [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: 01/31/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/14/2022]
Abstract
Retinoblastoma (RB) is the most common primary intraocular malignancy of childhood. It is known that the tumor microenvironment (TME) regulates tumorigenesis and metastasis. However, how the malignant progression in RB is determined by the heterogeneity of tumor cells and TME remains uncharacterized. Here, we conducted integrative single-cell transcriptome and whole-exome sequencing analysis of RB patients with detailed pathological and clinical measurements. By single-cell transcriptomic sequencing, we profiled around 70,000 cells from tumor samples of seven RB patients. We identified that the major cell types in RB were cone precursor-like (CP-like) and MKI67+ cone precursor (MKI67+ CP) cells. By integrating copy number variation (CNV) analysis, we found that RB samples had large clonal heterogeneity, where the malignant MKI67+ CP cells had significantly larger copy number changes. Enrichment analysis revealed that the conversion of CP-like to MKI67+ CP resulted in the loss of photoreceptor function and increased cell proliferation ability. The TME in RB was composed of tumor-associated macrophages (TAMs), astrocyte-like, and cancer-associated fibroblasts (CAFs). Particularly, during the invasion process, TAMs created an immunosuppressive environment, in which the proportion of TAMs decreased, M1-type macrophage was lost, and the TAMs-related immune functions were depressed. Finally, we identified that TAMs regulated tumor cells through GRN and MIF signaling pathways, while TAMs self-regulated through inhibition of CCL and GALECTIN signaling pathways during the invasion process. Altogether, our study creates a detailed transcriptomic map of RB with single-cell characterization of malignant phenotypes and provides novel molecular insights into the occurrence and progression of RB.
Collapse
|
14
|
Rozanska A, Cerna-Chavez R, Queen R, Collin J, Zerti D, Dorgau B, Beh CS, Davey T, Coxhead J, Hussain R, Al-Aama J, Steel DH, Benvenisty N, Armstrong L, Parulekar M, Lako M. pRB-Depleted Pluripotent Stem Cell Retinal Organoids Recapitulate Cell State Transitions of Retinoblastoma Development and Suggest an Important Role for pRB in Retinal Cell Differentiation. Stem Cells Transl Med 2022; 11:415-433. [PMID: 35325233 PMCID: PMC9052432 DOI: 10.1093/stcltm/szac008] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/19/2021] [Indexed: 11/12/2022] Open
Abstract
Retinoblastoma (Rb) is a childhood cancer of the developing retina, accounting for up to 17% of all tumors in infancy. To gain insights into the transcriptional events of cell state transitions during Rb development, we established 2 disease models via retinal organoid differentiation of a pRB (retinoblastoma protein)-depleted human embryonic stem cell line (RB1-null hESCs) and a pRB patient-specific induced pluripotent (iPSC) line harboring a RB1 biallelic mutation (c.2082delC). Both models were characterized by pRB depletion and accumulation of retinal progenitor cells at the expense of amacrine, horizontal and retinal ganglion cells, which suggests an important role for pRB in differentiation of these cell lineages. Importantly, a significant increase in the fraction of proliferating cone precursors (RXRγ+Ki67+) was observed in both pRB-depleted organoid models, which were defined as Rb-like clusters by single-cell RNA-Seq analysis. The pRB-depleted retinal organoids displayed similar features to Rb tumors, including mitochondrial cristae aberrations and rosette-like structures, and were able to undergo cell growth in an anchorage-independent manner, indicative of cell transformation in vitro. In both models, the Rb cones expressed retinal ganglion and horizontal cell markers, a novel finding, which could help to better characterize these tumors with possible therapeutic implications. Application of Melphalan, Topotecan, and TW-37 led to a significant reduction in the fraction of Rb proliferating cone precursors, validating the suitability of these in vitro models for testing novel therapeutics for Rb.
Collapse
Affiliation(s)
- Agata Rozanska
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Rachel Queen
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Darin Zerti
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Birthe Dorgau
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Chia Shyan Beh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tracey Davey
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jonathan Coxhead
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Rafiqul Hussain
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jumana Al-Aama
- Faculty of Medicine, King Abdulaziz University, Riyadh, Saudi Arabia
| | - David H Steel
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Manoj Parulekar
- Birmingham Women's and Children NHS Foundation Trust, Birmingham, UK
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
15
|
RB1-Negative Retinal Organoids Display Proliferation of Cone Photoreceptors and Loss of Retinal Differentiation. Cancers (Basel) 2022; 14:cancers14092166. [PMID: 35565295 PMCID: PMC9105736 DOI: 10.3390/cancers14092166] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/14/2022] [Accepted: 04/22/2022] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Retinoblastoma is a tumor of the eye’s retina, which is the very specialized tissue responsible for vision. In 98% of cases, the tumor is caused by inactivation of the RB1 gene. Due to lack of material and models, the understanding of RB1 mutations in tumor development is still unsatisfactory. We aimed to establish a human laboratory model for retinoblastoma. While differentiating stem cells with a mutation in RB1 into retina, we observed reduced differentiation potential but enhanced proliferation—general hallmarks of tumor development. The gene expression signature in the model resembled that of tumor material. This approach now enables research on retinoblastoma and probably therapy in the correct tissue, the human retina. Abstract Retinoblastoma is a tumor of the eye in children under the age of five caused by biallelic inactivation of the RB1 tumor suppressor gene in maturing retinal cells. Cancer models are essential for understanding tumor development and in preclinical research. Because of the complex organization of the human retina, such models were challenging to develop for retinoblastoma. Here, we present an organoid model based on differentiation of human embryonic stem cells into neural retina after inactivation of RB1 by CRISPR/Cas9 mutagenesis. Wildtype and RB1 heterozygous mutant retinal organoids were indistinguishable with respect to morphology, temporal development of retinal cell types and global mRNA expression. However, loss of pRB resulted in spatially disorganized organoids and aberrant differentiation, indicated by disintegration of organoids beyond day 130 of differentiation and depletion of most retinal cell types. Only cone photoreceptors were abundant and continued to proliferate, supporting these as candidate cells-of-origin for retinoblastoma. Transcriptome analysis of RB1 knockout organoids and primary retinoblastoma revealed gain of a retinoblastoma expression signature in the organoids, characterized by upregulation of RBL1 (p107), MDM2, DEK, SYK and HELLS. In addition, genes related to immune response and extracellular matrix were specifically upregulated in RB1-negative organoids. In vitro retinal organoids therefore display some features associated with retinoblastoma and, so far, represent the only valid human cancer model for the development of this disease.
Collapse
|
16
|
Liu J, Ottaviani D, Sefta M, Desbrousses C, Chapeaublanc E, Aschero R, Sirab N, Lubieniecki F, Lamas G, Tonon L, Dehainault C, Hua C, Fréneaux P, Reichman S, Karboul N, Biton A, Mirabal-Ortega L, Larcher M, Brulard C, Arrufat S, Nicolas A, Elarouci N, Popova T, Némati F, Decaudin D, Gentien D, Baulande S, Mariani O, Dufour F, Guibert S, Vallot C, Rouic LLL, Matet A, Desjardins L, Pascual-Pasto G, Suñol M, Catala-Mora J, Llano GC, Couturier J, Barillot E, Schaiquevich P, Gauthier-Villars M, Stoppa-Lyonnet D, Golmard L, Houdayer C, Brisse H, Bernard-Pierrot I, Letouzé E, Viari A, Saule S, Sastre-Garau X, Doz F, Carcaboso AM, Cassoux N, Pouponnot C, Goureau O, Chantada G, de Reyniès A, Aerts I, Radvanyi F. A high-risk retinoblastoma subtype with stemness features, dedifferentiated cone states and neuronal/ganglion cell gene expression. Nat Commun 2021; 12:5578. [PMID: 34552068 PMCID: PMC8458383 DOI: 10.1038/s41467-021-25792-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Retinoblastoma is the most frequent intraocular malignancy in children, originating from a maturing cone precursor in the developing retina. Little is known on the molecular basis underlying the biological and clinical behavior of this cancer. Here, using multi-omics data, we demonstrate the existence of two retinoblastoma subtypes. Subtype 1, of earlier onset, includes most of the heritable forms. It harbors few genetic alterations other than the initiating RB1 inactivation and corresponds to differentiated tumors expressing mature cone markers. By contrast, subtype 2 tumors harbor frequent recurrent genetic alterations including MYCN-amplification. They express markers of less differentiated cone together with neuronal/ganglion cell markers with marked inter- and intra-tumor heterogeneity. The cone dedifferentiation in subtype 2 is associated with stemness features including low immune and interferon response, E2F and MYC/MYCN activation and a higher propensity for metastasis. The recognition of these two subtypes, one maintaining a cone-differentiated state, and the other, more aggressive, associated with cone dedifferentiation and expression of neuronal markers, opens up important biological and clinical perspectives for retinoblastomas.
Collapse
Affiliation(s)
- Jing Liu
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Daniela Ottaviani
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.414531.60000 0001 0695 6255Precision Medicine, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Meriem Sefta
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Céline Desbrousses
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Elodie Chapeaublanc
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Rosario Aschero
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Nanor Sirab
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Fabiana Lubieniecki
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Gabriela Lamas
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Laurie Tonon
- grid.418116.b0000 0001 0200 3174Synergie Lyon Cancer, Plateforme de Bioinformatique “Gilles Thomas”, Centre Léon Bérard, 69008 Lyon, France
| | - Catherine Dehainault
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France
| | - Clément Hua
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Paul Fréneaux
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Sacha Reichman
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Narjesse Karboul
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Anne Biton
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U900, 75005 Paris, France ,Ecole des Mines ParisTech, 77305 Fontainebleau, France ,grid.428999.70000 0001 2353 6535Present Address: Institut Pasteur – Hub Bioinformatique et Biostatistique – C3BI, USR 3756 IP CNRS, 75015 Paris, France
| | - Liliana Mirabal-Ortega
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Magalie Larcher
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Céline Brulard
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.411777.30000 0004 1765 1563Present Address: INSERM U930, CHU Bretonneau, 37000 Tours, France
| | - Sandrine Arrufat
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - André Nicolas
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Nabila Elarouci
- grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Tatiana Popova
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Fariba Némati
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - Didier Decaudin
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - David Gentien
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - Sylvain Baulande
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, NGS Platform, 75005 Paris, France
| | - Odette Mariani
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Florent Dufour
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Sylvain Guibert
- grid.425132.3GeCo Genomics Consulting, Integragen, 91000 Evry, France
| | - Céline Vallot
- grid.425132.3GeCo Genomics Consulting, Integragen, 91000 Evry, France
| | - Livia Lumbroso-Le Rouic
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France
| | - Alexandre Matet
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Laurence Desjardins
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France
| | - Guillem Pascual-Pasto
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Mariona Suñol
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Department of Pathology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Jaume Catala-Mora
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Department of Ophthalmology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Genoveva Correa Llano
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Jérôme Couturier
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Emmanuel Barillot
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U900, 75005 Paris, France ,Ecole des Mines ParisTech, 77305 Fontainebleau, France
| | - Paula Schaiquevich
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina ,grid.423606.50000 0001 1945 2152National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Marion Gauthier-Villars
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Dominique Stoppa-Lyonnet
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Lisa Golmard
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Claude Houdayer
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France ,grid.41724.34Present Address: Department of Genetics, Rouen University Hospital, 76000 Rouen, France
| | - Hervé Brisse
- grid.418596.70000 0004 0639 6384Département d’Imagerie Médicale, Institut Curie, 75005 Paris, France
| | - Isabelle Bernard-Pierrot
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Eric Letouzé
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Universités, INSERM, 75006 Paris, France ,grid.508487.60000 0004 7885 7602Functional Genomics of Solid Tumors, équipe labellisée Ligue Contre le Cancer, Université de Paris, Université Paris 13, Paris, France
| | - Alain Viari
- grid.418116.b0000 0001 0200 3174Synergie Lyon Cancer, Plateforme de Bioinformatique “Gilles Thomas”, Centre Léon Bérard, 69008 Lyon, France
| | - Simon Saule
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Xavier Sastre-Garau
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.414145.10000 0004 1765 2136Present Address: Department of Pathology, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France
| | - François Doz
- grid.508487.60000 0004 7885 7602Université de Paris, Paris, France ,grid.418596.70000 0004 0639 6384SIREDO Center (Care, Innovation and Research in Pediatric Adolescent and Young Adult Oncology), Institut Curie, 75005 Paris, France
| | - Angel M. Carcaboso
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Nathalie Cassoux
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Celio Pouponnot
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Guillermo Chantada
- grid.414531.60000 0001 0695 6255Precision Medicine, Hospital J.P. Garrahan, Buenos Aires, Argentina ,grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain ,grid.423606.50000 0001 1945 2152National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Aurélien de Reyniès
- grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Isabelle Aerts
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.418596.70000 0004 0639 6384SIREDO Center (Care, Innovation and Research in Pediatric Adolescent and Young Adult Oncology), Institut Curie, 75005 Paris, France
| | - François Radvanyi
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| |
Collapse
|
17
|
Ying P, Huang C, Wang Y, Guo X, Cao Y, Zhang Y, Fu S, Chen L, Yi G, Fu M. Single-Cell RNA Sequencing of Retina:New Looks for Gene Marker and Old Diseases. Front Mol Biosci 2021; 8:699906. [PMID: 34395530 PMCID: PMC8362665 DOI: 10.3389/fmolb.2021.699906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/01/2021] [Indexed: 01/20/2023] Open
Abstract
The retina is composed of 11 types of cells, including neurons, glial cells and vascular bed cells. It contains five types of neurons, each with specific physiological, morphological, and molecular definitions. Currently, single-cell RNA sequencing (sRNA-seq) is emerging as one of the most powerful tools to reveal the complexity of the retina. The continuous discovery of retina-related gene targets plays an important role in helping us understand the nature of diseases. The revelation of new cell subpopulations can focus the occurrence and development of diseases on specific biological activities of specific cells. In addition, sRNA-seq performs high-throughput sequencing analysis of epigenetics, transcriptome and genome at the single-cell level, with the advantages of high-throughput and high-resolution. In this paper, we systematically review the development history of sRNA-seq technology, and summarize the new subtypes of retinal cells and some specific gene markers discovered by this technology. The progress in the diagnosis of retinal related diseases is also discussed.
Collapse
Affiliation(s)
- Peixi Ying
- The Second Clinical School, Southern Medical University, Guangzhou, China
| | - Chang Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yan Wang
- Department of Ophthalmology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Xi Guo
- Medical College of Rehabiliation, Southern Medical University, Guangzhou, China
| | - Yuchen Cao
- The Second Clinical School, Southern Medical University, Guangzhou, China
| | - Yuxi Zhang
- The Second Clinical School, Southern Medical University, Guangzhou, China
| | - Sheng Fu
- The University of South China, Hengyang, China
| | - Lin Chen
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Guoguo Yi
- Department of Ophthalmology, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Retinoblastoma from human stem cell-derived retinal organoids. Nat Commun 2021; 12:4535. [PMID: 34315877 PMCID: PMC8316454 DOI: 10.1038/s41467-021-24781-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
Retinoblastoma is a childhood cancer of the developing retina that initiates with biallelic inactivation of the RB1 gene. Children with germline mutations in RB1 have a high likelihood of developing retinoblastoma and other malignancies later in life. Genetically engineered mouse models of retinoblastoma share some similarities with human retinoblastoma but there are differences in their cellular differentiation. To develop a laboratory model of human retinoblastoma formation, we make induced pluripotent stem cells (iPSCs) from 15 participants with germline RB1 mutations. Each of the stem cell lines is validated, characterized and then differentiated into retina using a 3-dimensional organoid culture system. After 45 days in culture, the retinal organoids are dissociated and injected into the vitreous of eyes of immunocompromised mice to support retinoblastoma tumor growth. Retinoblastomas formed from retinal organoids made from patient-derived iPSCs have molecular, cellular and genomic features indistinguishable from human retinoblastomas. This model of human cancer based on patient-derived iPSCs with germline cancer predisposing mutations provides valuable insights into the cellular origins of this debilitating childhood disease as well as the mechanism of tumorigenesis following RB1 gene inactivation. Retinoblastoma is a heritable pediatric cancer driven by mutations in RB1. Here, the authors demonstrate the first patient derived model of retinoblastoma using iPSCs from patients with germline mutations in RB1.
Collapse
|
19
|
Sarver AL, Xie C, Riddle MJ, Forster CL, Wang X, Lu H, Wagner W, Tolar J, Hallstrom TC. Retinoblastoma tumor cell proliferation is negatively associated with an immune gene expression signature and increased immune cells. J Transl Med 2021; 101:701-718. [PMID: 33658609 DOI: 10.1038/s41374-021-00573-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/30/2022] Open
Abstract
This study focuses on gene expression differences between early retinal states that ultimately lead to normal development, late onset retinoblastoma, or rapid bilateral retinoblastoma tumors. The late-onset and early-onset retinoblastoma tumor cells are remarkably similar to normally proliferating retinal progenitor cells, but they fail to properly express differentiation markers associated with normal development. Further, early-onset retinoblastoma tumor cells express a robust immune gene expression signature followed by accumulation of dendritic, monocyte, macrophage, and T-lymphocyte cells in the retinoblastoma tumors. This characteristic was not shared by either normal retinae or late-onset retinoblastomas. Comparison of our data with other human and mouse retinoblastoma tumor gene expression significantly confirmed, that the immune signature is present in tumors from each species. Strikingly, we observed that the immune signature in both mouse and human tumors was most highly evident in those with the lowest proliferative capacity. We directly assessed this relationship in human retinoblastoma tumors by co-analyzing proliferation and immune cell recruitment by immunohistochemistry, uncovering a significant inverse relationship between increased immune-cell infiltration in tumors and reduced tumor cell proliferation. Directly inhibiting proliferation with a PI3K/mTOR inhibitor significantly increased the number of CD45+ immune cells in the retina. This work establishes an in vivo model for the rapid recruitment of immune cells to tumorigenic neural tissue.
Collapse
Affiliation(s)
- Aaron L Sarver
- Institute for Health Informatics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Chencheng Xie
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Megan J Riddle
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Colleen L Forster
- BioNet, Academic Health Center, University of Minnesota, Minneapolis, MN, USA
| | - Xiaohong Wang
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Huarui Lu
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Wyatt Wagner
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Jakub Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Timothy C Hallstrom
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
20
|
Saengwimol D, Chittavanich P, Laosillapacharoen N, Srimongkol A, Chaitankar V, Rojanaporn D, Aroonroch R, Suktitipat B, Saisawang C, Svasti S, Hongeng S, Kaewkhaw R. Silencing of the Long Noncoding RNA MYCNOS1 Suppresses Activity of MYCN-Amplified Retinoblastoma Without RB1 Mutation. Invest Ophthalmol Vis Sci 2021; 61:8. [PMID: 33270844 PMCID: PMC7718827 DOI: 10.1167/iovs.61.14.8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose MYCNOS (MYCN opposite strand) is co-amplified with MYCN in pediatric cancers, including retinoblastoma. MYCNOS encodes several RNA variants whose functions have not been elucidated in retinoblastoma. Thus, we attempted to identify MYCNOS variants in retinoblastoma and aimed to decipher the role of MYCNOS variant 1 (MYCNOS1) on the activity of MYCN-amplified retinoblastoma. Methods The profiles of MYCNOS variants and MYCN status were determined in 17 retinoblastoma tissues, cell lines, retinas, and retinal organoids. A functional study of MYCNOS1 expression was conducted in patient-derived tumor cells and in retinoblastoma cell lines via short hairpin RNA-mediated gene silencing. We carried out MYCN expression, cell viability, cell cycle, apoptosis, soft agar colony formation, and transwell assays to examine the role of MYCNOS1 in MYCN and cell behaviors. We analyzed a transcriptome of MYCN-amplified retinoblastoma cells deficient for MYCNOS1 and, finally, tested the responses of these cells to chemotherapeutic agents. Results Expression of MYCNOS1 was associated with the expression and copy number of MYCN. Knockdown of MYCNOS1 caused instability of the MYCN protein, leading to cell cycle arrest and impaired proliferation and chemotaxis-directed migration in MYCN-amplified retinoblastoma cells in which RB1 was intact. MYCNOS1 expression was associated with gene signatures of photoreceptor cells and epithelial–mesenchymal transition. MYCNOS1 silencing enhanced the response of retinoblastoma cells to topotecan but not carboplatin. Conclusions MYCNOS1 supports progression of retinoblastoma. Inhibition of MYCNOS1 expression may be necessary to suppress MYCN activity when treating MYCN-amplified cancers without RB1 mutation.
Collapse
Affiliation(s)
- Duangporn Saengwimol
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pamorn Chittavanich
- Section of Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Natanan Laosillapacharoen
- Section of Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Atthapol Srimongkol
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Vijender Chaitankar
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Duangnate Rojanaporn
- Department of Ophthalmology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rangsima Aroonroch
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Bhoom Suktitipat
- Department of Biochemistry, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom, Thailand
| | - Chonticha Saisawang
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Saovaros Svasti
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand.,Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rossukon Kaewkhaw
- Section of Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
21
|
Collin J, Queen R, Zerti D, Steel DH, Bowen C, Parulekar M, Lako M. Dissecting the Transcriptional and Chromatin Accessibility Heterogeneity of Proliferating Cone Precursors in Human Retinoblastoma Tumors by Single Cell Sequencing-Opening Pathways to New Therapeutic Strategies? Invest Ophthalmol Vis Sci 2021; 62:18. [PMID: 34003213 PMCID: PMC8132003 DOI: 10.1167/iovs.62.6.18] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/18/2021] [Indexed: 01/03/2023] Open
Abstract
Purpose Retinoblastoma (Rb) is a malignant neoplasm arising during retinal development from mutations in the RB1 gene. Loss or inactivation of both copies of RB1 results in initiation of retinoblastoma tumors; however, additional genetic changes are needed for the continued growth and spread of the tumor. Ex vivo research has shown that in humans, retinoblastoma may initiate from RB1-depleted cone precursors. Notwithstanding, it has not been possible to assess the full spectrum of clonal types within the tumor itself in vivo and the molecular changes occurring at the cells of origin, enabling their malignant conversion. To overcome these challenges, we have performed the first single cell (sc) RNA- and ATAC-Seq analyses of primary tumor tissues, enabling us to dissect the transcriptional and chromatin accessibility heterogeneity of proliferating cone precursors in human Rb tumors. Methods Two Rb tumors each characterized by two pathogenic RB1 mutations were dissociated to single cells and subjected to scRNA-Seq and scATAC-Seq using the 10× Genomics platform. In addition, nine human embryonic and fetal retina samples were dissociated to single cells and subjected to scRNA- and ATAC-Seq analyses. The scRNA- and ATAC-Seq data were embedded using Uniform Manifold Approximation and Projection and clustered with Seurat graph-based clustering. Integrated scATAC-Seq analysis of Rb tumors and human embryonic/fetal retina samples was performed to identify Rb cone enriched subclusters. Pseudo time analysis of proliferating cones in the Rb samples was performed with Monocle. Ingenuity Pathway Analysis was used to identify the signaling pathway and upstream regulators in the Rb cone-enriched subclusters. Results Our single cell analyses revealed the predominant presence of cone precursors at different stages of the cell cycle in the Rb tumors and among those identified the G2/M subset as the cell type of origin. scATAC-Seq analysis identified two Rb enriched cone subclusters, each characterized by activation of different upstream regulators and signaling pathways, enabling proliferating cone precursors to escape cell cycle arrest and/or apoptosis. Conclusions Our study provides evidence of Rb tumor heterogeneity and defines molecular pathways that can be targeted to define new treatment strategies.
Collapse
Affiliation(s)
- Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Rachel Queen
- Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Darin Zerti
- Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - David H Steel
- Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Claire Bowen
- Birmingham Women's and Children NHS Foundation Trust, Birmingham, United Kingdom
| | - Manoj Parulekar
- Birmingham Women's and Children NHS Foundation Trust, Birmingham, United Kingdom
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| |
Collapse
|
22
|
He Z, Pan Y, Shao F, Wang H. Identifying Differentially Expressed Genes of Zero Inflated Single Cell RNA Sequencing Data Using Mixed Model Score Tests. Front Genet 2021; 12:616686. [PMID: 33613638 PMCID: PMC7894898 DOI: 10.3389/fgene.2021.616686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Single cell RNA sequencing (scRNA-seq) allows quantitative measurement and comparison of gene expression at the resolution of single cells. Ignoring the batch effects and zero inflation of scRNA-seq data, many proposed differentially expressed (DE) methods might generate bias. We propose a method, single cell mixed model score tests (scMMSTs), to efficiently identify DE genes of scRNA-seq data with batch effects using the generalized linear mixed model (GLMM). scMMSTs treat the batch effect as a random effect. For zero inflation, scMMSTs use a weighting strategy to calculate observational weights for counts independently under zero-inflated and zero-truncated distributions. Counts data with calculated weights were subsequently analyzed using weighted GLMMs. The theoretical null distributions of the score statistics were constructed by mixed Chi-square distributions. Intensive simulations and two real datasets were used to compare edgeR-zinbwave, DESeq2-zinbwave, and scMMSTs. Our study demonstrates that scMMSTs, as supplement to standard methods, are advantageous to define DE genes of zero-inflated scRNA-seq data with batch effects.
Collapse
Affiliation(s)
- Zhiqiang He
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yueyun Pan
- First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Fang Shao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hui Wang
- Department of Maternal and Child Health, School of Public Health, Peking University Health Science Center, Beijing, China
| |
Collapse
|
23
|
Sheppard HE, Dall’Agnese A, Park WD, Shamim MH, Dubrulle J, Johnson HL, Stossi F, Cogswell P, Sommer J, Levy J, Sharifnia T, Wawer MJ, Nabet B, Gray NS, Clemons PA, Schreiber SL, Workman P, Young RA, Lin CY. Targeted brachyury degradation disrupts a highly specific autoregulatory program controlling chordoma cell identity. CELL REPORTS MEDICINE 2021; 2:100188. [PMID: 33521702 PMCID: PMC7817874 DOI: 10.1016/j.xcrm.2020.100188] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 08/14/2020] [Accepted: 12/17/2020] [Indexed: 12/31/2022]
Abstract
Chordomas are rare spinal tumors addicted to expression of the developmental transcription factor brachyury. In chordomas, brachyury is super-enhancer associated and preferentially downregulated by pharmacologic transcriptional CDK inhibition, leading to cell death. To understand the underlying basis of this sensitivity, we dissect the brachyury transcription regulatory network and compare the consequences of brachyury degradation with transcriptional CDK inhibition. Brachyury defines the chordoma super-enhancer landscape and autoregulates through binding its super-enhancer, and its locus forms a transcriptional condensate. Transcriptional CDK inhibition and brachyury degradation disrupt brachyury autoregulation, leading to loss of its transcriptional condensate and transcriptional program. Compared with transcriptional CDK inhibition, which globally downregulates transcription, leading to cell death, brachyury degradation is much more selective, inducing senescence and sensitizing cells to anti-apoptotic inhibition. These data suggest that brachyury downregulation is a core tenet of transcriptional CDK inhibition and motivates developing strategies to target brachyury and its autoregulatory feedback loop. Brachyury defines the chordoma super-enhancer landscape Brachyury autoregulates through a transcriptional condensate CDK7/12/13i and brachyury degradation target the brachyury transcriptional condensate Brachyury degradation inhibits chordoma identity genes and induces senescence
Collapse
Affiliation(s)
- Hadley E. Sheppard
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Woojun D. Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - M. Hamza Shamim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julien Dubrulle
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hannah L. Johnson
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fabio Stossi
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Joan Levy
- Chordoma Foundation, Durham, NC 27713, USA
| | - Tanaz Sharifnia
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | | | - Behnam Nabet
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nathanael S. Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Paul A. Clemons
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Stuart L. Schreiber
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| | - Richard A. Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charles Y. Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Therapeutic Innovation Center, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Corresponding author
| |
Collapse
|
24
|
Guihurt Santiago J, Burgos-Tirado N, Lafontaine DD, Mendoza Sierra JC, Camacho RH, Vecchini Rodríguez CM, Morales-Tirado V, Flores-Otero J. Adhesion G protein-coupled receptor, ELTD1, is a potential therapeutic target for retinoblastoma migration and invasion. BMC Cancer 2021; 21:53. [PMID: 33430814 PMCID: PMC7802354 DOI: 10.1186/s12885-020-07768-3] [Citation(s) in RCA: 7] [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: 07/05/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Prognosis for pediatric metastatic Retinoblastoma (Rb) is poor and current therapies are limited by high systemic toxicity rates and insufficient therapeutic efficacy for metastatic Rb. Tumor dissemination to the brain is promoted by the heterogeneous adhesive and invasive properties of Rb cells within the tumor. In this study we evaluate, for the first time, the expression, and roles of the ELTD1 and GPR125 adhesion G protein-coupled receptors (GPCRs) in Rb cell migration, viability and invasion. METHODS We characterized the RNA expression of adhesion-GPCRs in 64 Rb tumors compared to 11 fetal retinas using the database from the Childhood Solid Tumor Network from St Jude Children's Research Hospital. The role of ELTD1 and GPR125 in Rb were investigated ex vivo by microarray analysis, in vitro by cell viability, Western blot and migration assays, in addition to imaging of the subcellular localization of the GPCRs. To elucidate their role in vivo we utilized siRNA technology in an established Rb orthotopic xenograft murine model. RESULTS Our investigation demonstrates, for the first time, that ELTD1 but not GPR125, is significantly increased in Rb tumors compared to fetal retinas. We utilized established the Rb cell lines Y79 and Weri-Rb-1, which represent an aggressive, metastatic, and non-metastatic phenotype, respectively, for the in vitro analyses. The studies demonstrated that ELTD1 is enriched in Weri-Rb-1 cells, while GPR125 is enriched in Y79 cells. The measured differences extended to their subcellular localization as ELTD1 labeling displayed punctate clusters in cell-to-cell adhesion sites of Weri-Rb-1 cells, while GPR125 displayed a polarized distribution in Y79 cells. Lastly, we demonstrated the lack of both adhesion receptors does not affect Rb cell viability, yet inhibition of ELTD1 decreases Y79 cell migration in vitro and invasion in vivo. CONCLUSION Taken together, our data suggest that ELTD1, is a potential target to prevent extraocular Rb. The results within establish ELTD1 as a potential therapeutic target for metastatic Rb.
Collapse
Affiliation(s)
- Jonathan Guihurt Santiago
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- University of Puerto Rico, Rio Piedras Campus, Rio Piedras, Puerto Rico
- Present address: Debusk College of Osteopathic Medicine at Lincoln Memorial University, Harrogate, TN USA
| | - Neikelyn Burgos-Tirado
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- University of Puerto Rico, Rio Piedras Campus, Rio Piedras, Puerto Rico
- Present address: Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI USA
| | - Daniella Dorta Lafontaine
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- University of Puerto Rico, Rio Piedras Campus, Rio Piedras, Puerto Rico
- Present address: Central University of the Caribbean of Puerto Rico, Bayamon, Puerto Rico
| | - José C. Mendoza Sierra
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- University of Puerto Rico, Rio Piedras Campus, Rio Piedras, Puerto Rico
- University of Medicine and Health Sciences, New York, USA
| | - Roberto Herrera Camacho
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- Current affiliation: Ponce Health Sciences University, Ponce, Puerto Rico
| | - Clara M. Vecchini Rodríguez
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- Department of Anatomy and Neurobiology, University of Puerto Rico, Medical Sciences Campus, PO Box 365067, San Juan, 00936-5067 Puerto Rico
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| | - Vanessa Morales-Tirado
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN USA
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN USA
- Present address: AbbVie Bioresearch Center, Worcester, MA USA
| | - Jacqueline Flores-Otero
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- Department of Anatomy and Neurobiology, University of Puerto Rico, Medical Sciences Campus, PO Box 365067, San Juan, 00936-5067 Puerto Rico
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| |
Collapse
|
25
|
Human embryonic stem cell-derived organoid retinoblastoma reveals a cancerous origin. Proc Natl Acad Sci U S A 2020; 117:33628-33638. [PMID: 33318192 PMCID: PMC7776986 DOI: 10.1073/pnas.2011780117] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
As a genetic malignancy, retinoblastoma (Rb) is caused by RB1 mutations; however, its developmental origin and drug agents for human Rb remain largely unexplored. Here we describe an innovative Rb organoid model derived from human embryonic stem cells with a biallelic mutagenesis of the RB1 gene. We identify tumorigenic growth in the Rb organoids, as well as properties consistent with human primary Rb. We confirm that the Rb cell of origin stemmed from ARR3+ maturing cone precursor cells and SYK inhibitors displaying a significant therapeutic response. Our elegant in-dish Rb organoid model can be used to efficiently and effectively dissect the origin of Rb and mechanisms of Rb tumorigenesis, as well as screen novel therapies. Retinoblastoma (Rb) is the most prevalent intraocular malignancy in children, with a worldwide survival rate <30%. We have developed a cancerous model of Rb in retinal organoids derived from genetically engineered human embryonic stem cells (hESCs) with a biallelic mutagenesis of the RB1 gene. These organoid Rbs exhibit properties highly consistent with Rb tumorigenesis, transcriptome, and genome-wide methylation. Single-cell sequencing analysis suggests that Rb originated from ARR3-positive maturing cone precursors during development, which was further validated by immunostaining. Notably, we found that the PI3K-Akt pathway was aberrantly deregulated and its activator spleen tyrosine kinase (SYK) was significantly up-regulated. In addition, SYK inhibitors led to remarkable cell apoptosis in cancerous organoids. In conclusion, we have established an organoid Rb model derived from genetically engineered hESCs in a dish that has enabled us to trace the cell of origin and to test novel candidate therapeutic agents for human Rb, shedding light on the development and therapeutics of other malignancies.
Collapse
|
26
|
Lee C, Kim JK. Chromatin regulators in retinoblastoma: Biological roles and therapeutic applications. J Cell Physiol 2020; 236:2318-2332. [PMID: 32840881 PMCID: PMC7891620 DOI: 10.1002/jcp.30022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022]
Abstract
Retinoblastoma (RB) is a pediatric ocular tumor mostly occurring due to the biallelic loss of RB1 gene in the developing retina. Early studies of genomic aberrations in RB have provided a valuable insight into how RB can progress following the tumor-initiating RB1 mutations and have established a notion that inactivation of RB1 gene is critical to initiate RB but this causative genetic lesion alone is not sufficient for malignant progression. With the advent of high-throughput sequencing technologies, we now have access to the comprehensive genomic and epigenetic landscape of RB and have come to appreciate that RB tumorigenesis requires both genetic and epigenetic alterations that might be directly or indirectly driven by RB1 loss. This integrative perspective on RB tumorigenesis has inspired research efforts to better understand the types and functions of epigenetic mechanisms contributing to RB development, leading to the identification of multiple epigenetic regulators misregulated in RB in recent years. A complete understanding of the intricate network of genetic and epigenetic factors in modulation of gene expression during RB tumorigenesis remains a major challenge but would be crucial to translate these findings into therapeutic interventions. In this review, we will provide an overview of chromatin regulators identified to be misregulated in human RB among the numerous epigenetic factors implicated in RB development. For a subset of these chromatin regulators, recent findings on their functions in RB development and potential therapeutic applications are discussed.
Collapse
Affiliation(s)
- Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jong Kyong Kim
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Retinoblastoma: Etiology, Modeling, and Treatment. Cancers (Basel) 2020; 12:cancers12082304. [PMID: 32824373 PMCID: PMC7465685 DOI: 10.3390/cancers12082304] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/19/2022] Open
Abstract
Retinoblastoma is a retinal cancer that is initiated in response to biallelic loss of RB1 in almost all cases, together with other genetic/epigenetic changes culminating in the development of cancer. RB1 deficiency makes the retinoblastoma cell-of-origin extremely susceptible to cancerous transformation, and the tumor cell-of-origin appears to depend on the developmental stage and species. These are important to establish reliable preclinical models to study the disease and develop therapies. Although retinoblastoma is the most curable pediatric cancer with a high survival rate, advanced tumors limit globe salvage and are often associated with high-risk histopathological features predictive of dissemination. The advent of chemotherapy has improved treatment outcomes, which is effective for globe preservation with new routes of targeted drug delivery. However, molecularly targeted therapeutics with more effectiveness and less toxicity are needed. Here, we review the current knowledge concerning retinoblastoma genesis with particular attention to the genomic and transcriptomic landscapes with correlations to clinicopathological characteristics, as well as the retinoblastoma cell-of-origin and current disease models. We further discuss current treatments, clinicopathological correlations, which assist in guiding treatment and may facilitate globe preservation, and finally we discuss targeted therapeutics for future treatments.
Collapse
|
28
|
Alvarez-Suarez DE, Tovar H, Hernández-Lemus E, Orjuela M, Sadowinski-Pine S, Cabrera-Muñoz L, Camacho J, Favari L, Hernández-Angeles A, Ponce-Castañeda MV. Discovery of a transcriptomic core of genes shared in 8 primary retinoblastoma with a novel detection score analysis. J Cancer Res Clin Oncol 2020; 146:2029-2040. [PMID: 32474753 DOI: 10.1007/s00432-020-03266-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/14/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE Expression microarrays are powerful technology that allows large-scale analysis of RNA profiles in a tissue; these platforms include underexploited detection scores outputs. We developed an algorithm using the detection score, to generate a detection profile of shared elements in retinoblastoma as well as to determine its transcriptomic size and structure. METHODS We analyzed eight briefly cultured primary retinoblastomas with the Human transcriptome array 2.0 (HTA2.0). Transcripts and genes detection scores were determined using the Detection Above Background algorithm (DABG). We used unsupervised and supervised computational tools to analyze detected and undetected elements; WebGestalt was used to explore functions encoded by genes in relevant clusters and performed experimental validation. RESULTS We found a core cluster with 7,513 genes detected and shared by all samples, 4,321 genes in a cluster that was commonly absent, and 7,681 genes variably detected across the samples accounting for tumor heterogeneity. Relevant pathways identified in the core cluster relate to cell cycle, RNA transport, and DNA replication. We performed a kinome analysis of the core cluster and found 4 potential therapeutic kinase targets. Through analysis of the variably detected genes, we discovered 123 differentially expressed transcripts between bilateral and unilateral cases. CONCLUSIONS This novel analytical approach allowed determining the retinoblastoma transcriptomic size, a shared active transcriptomic core among the samples, potential therapeutic target kinases shared by all samples, transcripts related to inter tumor heterogeneity, and to determine transcriptomic profiles without the need of control tissues. This approach is useful to analyze other cancer or tissue types.
Collapse
Affiliation(s)
- Diana E Alvarez-Suarez
- Medical Research Unit in Infectious Diseases, Hospital de Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Pharmacology Department, CINVESTAV, Mexico City, Mexico
| | - Hugo Tovar
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| | - Manuela Orjuela
- Epidemiology Department, Columbia University, Columbia, NY, USA
| | - Stanislaw Sadowinski-Pine
- Pathology Department, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City, Mexico
| | - Lourdes Cabrera-Muñoz
- Pathology Department, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City, Mexico
| | | | | | - Adriana Hernández-Angeles
- Medical Research Unit in Infectious Diseases, Hospital de Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - M Verónica Ponce-Castañeda
- Medical Research Unit in Infectious Diseases, Hospital de Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| |
Collapse
|
29
|
Analysis of the p53 pathway in peripheral blood of retinoblastoma patients; potential biomarkers. PLoS One 2020; 15:e0234337. [PMID: 32502182 PMCID: PMC7274427 DOI: 10.1371/journal.pone.0234337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Loss of retinoblastoma (RB) function in the cone cells during retina development is necessary but not sufficient for retinoblastoma development. It has been reported that in the absence of RB activity, a retinoma is generated, and the onset of retina cancer occurs until the p53 pathway is altered. Unlike other types of cancer, in retinoblastoma the p53 tumour suppressor is mostly wild type, although its two primary regulators, MDMX and MDM2, are commonly dysregulated. A mutated RB form is inherited in around 35% of the cases, but normally two, somatic mutations are needed to alter the RB function. Here we investigated the mRNA levels of RB, p53, MDMX and MDM2 in peripheral blood samples of retinoblastoma patients to monitor the pathway status of p53 in somatic cells. We sought to investigate the involvement of these genes in the development of retina cancer, with the aim of identifying biomarkers for early diagnosis of this disease.
Collapse
|
30
|
Cao M, Wang S, Zou J, Wang W. Bioinformatics analyses of retinoblastoma reveal the retinoblastoma progression subtypes. PeerJ 2020; 8:e8873. [PMID: 32509443 PMCID: PMC7246025 DOI: 10.7717/peerj.8873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Retinoblastoma (RB) is one common pediatric malignant tumor with dismal outcomes. Heterogeneity of RB and subtypes of RB were identified but the association between the subtypes of RB and RB progression have not been fully investigated. Methods Four public datasets were downloaded from Gene expression omnibus and normalization was performed to remove batch effect. Two public datasets were explored to obtain the RB progression gene signatures by differentially expression analysis while another two datasets were iterated for RB subtypes identification using consensus clustering. After the RB progressive subtype gene signatures were identified, we tested the diagnostic capacity of these gene signatures by receiver operation curve. Results Three hundreds and forty six genes that were enriched in cell cycle were identified as the progression signature in RB from two independent datasets. Four subtypes of RB were stratified by consensus clustering. A total of 21 genes from RB progression signature were differentially expressed between RB subtypes. One subtype with low expression cell division genes have less progression of all four subtypes. A panel of five RB subtype genes (CLUL1, CNGB1, ROM1, LRRC39 and RDH12) predict progression of RB. Conclusion Retinoblastoma is a highly heterogeneous tumor and the level of cell cycle related gene expression is associated with RB progression. A subpopulation of RB with high expression of visual perception has less progressive features. LRRC39 is potentially the RB progression subtype biomarker.
Collapse
Affiliation(s)
- Manjing Cao
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Sha Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China
| | - Jing Zou
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China
| | - Wanpeng Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China
| |
Collapse
|
31
|
Tian R, Zou H, Wang LF, Song MJ, Liu L, Zhang H. Identification of microRNA-mRNA regulatory networks and pathways related to retinoblastoma across human and mouse. Int J Ophthalmol 2020; 13:535-544. [PMID: 32399402 PMCID: PMC7137714 DOI: 10.18240/ijo.2020.04.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
AIM To explore the mRNA and pathways related to retinoblastoma (RB) genesis and development. METHODS Microarray datasets GSE29683 (human) and GSE29685 (mouse) were downloaded from NCBI GEO database. Homologous genes between the two species were identified using WGCNA, followed by protein-protein interaction (PPI) network construction and gene enrichment analysis. Disease-related miRNAs and pathways were retrieved from miR2Disease database and Comparative Toxicogenomics Database (CTD), respectively. RESULTS A total of 352 homologous genes were identified. Two pathways including "cell cycle" and "pathway in cancer" in CTD and enrichment analysis were identified and seven miRNAs (including hsa-miR-373, hsa-miR-34a, hsa-miR-129, hsa-miR-494, hsa-miR-503, hsa-let-7 and hsa-miR-518c) were associated with RB. miRNAs modulate "cell cycle" and "pathway in cancer" pathways via regulating 13 genes (including CCND1, CDC25C, E2F2, CDKN2D and TGFB2). CONCLUSION These results suggest that these miRNAs play crucial roles in RB genesis through "cell cycle" and "pathway in cancer" pathways by regulating their targets including CCND1, CDC25C, E2F2 and CDKN2D.
Collapse
Affiliation(s)
- Rui Tian
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - He Zou
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Lu-Fei Wang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Mei-Jiao Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Lu Liu
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| | - Hui Zhang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, Jilin Province, China
| |
Collapse
|
32
|
Zheng C, Schneider JW, Hsieh J. Role of RB1 in human embryonic stem cell-derived retinal organoids. Dev Biol 2020; 462:197-207. [PMID: 32197890 DOI: 10.1016/j.ydbio.2020.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 02/21/2020] [Accepted: 03/11/2020] [Indexed: 01/09/2023]
Abstract
Three-dimensional (3D) organoid models derived from human pluripotent stem cells provide a platform for studying human development and understanding disease mechanisms. Most studies that examine biallelic inactivation of the cell cycle regulator Retinoblastoma 1 (RB1) and the link to retinoblastoma is in mice, however, less is known regarding the pathophysiological role of RB1 during human retinal development. To study the role of RB1 in early human retinal development and tumor formation, we generated retinal organoids from CRISPR/Cas9-derived RB1-null human embryonic stem cells (hESCs). We showed that RB is abundantly expressed in retinal progenitor cells in retinal organoids and loss of RB1 promotes S-phase entry. Furthermore, loss of RB1 resulted in widespread apoptosis and reduced the number of photoreceptor, ganglion, and bipolar cells. Interestingly, RB1 mutation in retinal organoids did not result in retinoblastoma formation in vitro or in the vitreous body of NOD/SCID immunodeficient mice. Together, our work identifies a crucial function for RB1 in human retinal development and suggests that RB1 deletion alone is not sufficient for tumor development, at least in human retinal organoids.
Collapse
Affiliation(s)
- Canbin Zheng
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Orthopedic and Microsurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, GD, 510080, China
| | - Jay W Schneider
- Wanek Program for Hypoplastic Left Heart Syndrome, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Jenny Hsieh
- Department of Biology and Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, 78249, USA.
| |
Collapse
|
33
|
Naert T, Dimitrakopoulou D, Tulkens D, Demuynck S, Carron M, Noelanders R, Eeckhout L, Van Isterdael G, Deforce D, Vanhove C, Van Dorpe J, Creytens D, Vleminckx K. RBL1 (p107) functions as tumor suppressor in glioblastoma and small-cell pancreatic neuroendocrine carcinoma in Xenopus tropicalis. Oncogene 2020; 39:2692-2706. [PMID: 32001819 DOI: 10.1038/s41388-020-1173-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 11/09/2022]
Abstract
Alterations of the retinoblastoma and/or the p53 signaling network are associated with specific cancers such as high-grade astrocytoma/glioblastoma, small-cell lung cancer (SCLC), choroid plexus tumors, and small-cell pancreatic neuroendocrine carcinoma (SC-PaNEC). However, the intricate functional redundancy between RB1 and the related pocket proteins RBL1/p107 and RBL2/p130 in suppressing tumorigenesis remains poorly understood. Here we performed lineage-restricted parallel inactivation of rb1 and rbl1 by multiplex CRISPR/Cas9 genome editing in the true diploid Xenopus tropicalis to gain insight into this in vivo redundancy. We show that while rb1 inactivation is sufficient to induce choroid plexus papilloma, combined rb1 and rbl1 inactivation is required and sufficient to drive SC-PaNEC, retinoblastoma and astrocytoma. Further, using a novel Li-Fraumeni syndrome-mimicking tp53 mutant X. tropicalis line, we demonstrate increased malignancy of rb1/rbl1-mutant glioma towards glioblastoma upon concomitant inactivation of tp53. Interestingly, although clinical SC-PaNEC samples are characterized by abnormal p53 expression or localization, in the current experimental models, the tp53 status had little effect on the establishment and growth of SC-PaNEC, but may rather be essential for maintaining chromosomal stability. SCLC was only rarely observed in our experimental setup, indicating requirement of additional or alternative oncogenic insults. In conclusion, we used CRISPR/Cas9 to delineate the tumor suppressor properties of Rbl1, generating new insights in the functional redundancy within the retinoblastoma protein family in suppressing neuroendocrine pancreatic cancer and glioma/glioblastoma.
Collapse
Affiliation(s)
- Thomas Naert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Dionysia Dimitrakopoulou
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Dieter Tulkens
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Suzan Demuynck
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marjolein Carron
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Rivka Noelanders
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Liza Eeckhout
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Dieter Deforce
- Laboratory for Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Cancer Research Institute Ghent, Ghent, Belgium
- Infinity lab, Ghent University Hospital, Ghent, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Pathology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - David Creytens
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Pathology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Kris Vleminckx
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent, Ghent, Belgium.
- Center for Medical Genetics, Ghent University, Ghent, Belgium.
| |
Collapse
|
34
|
Kim JK, Kan G, Mao Y, Wu Z, Tan X, He H, Lee C. UHRF1 downmodulation enhances antitumor effects of histone deacetylase inhibitors in retinoblastoma by augmenting oxidative stress-mediated apoptosis. Mol Oncol 2019; 14:329-346. [PMID: 31782885 PMCID: PMC6998393 DOI: 10.1002/1878-0261.12607] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/18/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022] Open
Abstract
Identification of new genetic pathways or molecular targets that sensitize cancer cells to chemotherapeutic drugs may improve the efficacy of current chemotherapy. Here, we report that downmodulation of UHRF1 (ubiquitin-like with PHD and RING finger domains 1) in retinoblastoma (RB) cells increases the sensitivity to histone deacetylase (HDAC) inhibitors, augmenting apoptotic cell death. We found that UHRF1 depletion downregulates two redox-responsive genes GSTA4 (glutathione S-transferase α4) and TXN2 (thioredoxin-2) in RB cells, and increases the basal level of intracellular oxidative stress. Antioxidant treatment significantly reduced both basal and HDAC inhibitor-induced DNA damage and apoptosis in UHRF1-depleted cells. Knockdown of GSTA4 or TXN2 sensitized RB cells to HDAC inhibitors, demonstrating that GSTA4 and TXN2 play key roles in redox homeostasis in RB cells and the susceptibility to HDAC inhibitor treatment upon UHRF1 depletion. In human primary RB, GSTA4 and TXN2 proteins were found to be mostly elevated along with high UHRF1 expression. In addition to augmentation of apoptosis in UHRF1-depleted RB cells, we also show that UHRF1 downmodulation derepresses the expression of photoreceptor-specific genes in RB cells in cooperation with a HDAC inhibitor MS-275 and promotes neuron-like differentiation. However, further investigation revealed that the enhanced growth-inhibitory effects of MS-275 in UHRF1-depleted cells were still mainly due to robust apoptosis induction rather than differentiation-mediated growth arrest. Consistent with our findings, UHRF1 depletion in RB cells increased the therapeutic efficacy of MS-275 in murine orthotopic xenografts. These results provide a novel basis for potential benefits of UHRF1 targeting for RB treatment.
Collapse
Affiliation(s)
- Jong Kyong Kim
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Guangyan Kan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yu Mao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhixuan Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xionghong Tan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Heng He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Munier FL, Beck-Popovic M, Chantada GL, Cobrinik D, Kivelä TT, Lohmann D, Maeder P, Moll AC, Carcaboso AM, Moulin A, Schaiquevich P, Bergin C, Dyson PJ, Houghton S, Puccinelli F, Vial Y, Gaillard MC, Stathopoulos C. Conservative management of retinoblastoma: Challenging orthodoxy without compromising the state of metastatic grace. "Alive, with good vision and no comorbidity". Prog Retin Eye Res 2019; 73:100764. [PMID: 31173880 DOI: 10.1016/j.preteyeres.2019.05.005] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/25/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Retinoblastoma is lethal by metastasis if left untreated, so the primary goal of therapy is to preserve life, with ocular survival, visual preservation and quality of life as secondary aims. Historically, enucleation was the first successful therapeutic approach to decrease mortality, followed over 100 years ago by the first eye salvage attempts with radiotherapy. This led to the empiric delineation of a window for conservative management subject to a "state of metastatic grace" never to be violated. Over the last two decades, conservative management of retinoblastoma witnessed an impressive acceleration of improvements, culminating in two major paradigm shifts in therapeutic strategy. Firstly, the introduction of systemic chemotherapy and focal treatments in the late 1990s enabled radiotherapy to be progressively abandoned. Around 10 years later, the advent of chemotherapy in situ, with the capitalization of new routes of targeted drug delivery, namely intra-arterial, intravitreal and now intracameral injections, allowed significant increase in eye preservation rate, definitive eradication of radiotherapy and reduction of systemic chemotherapy. Here we intend to review the relevant knowledge susceptible to improve the conservative management of retinoblastoma in compliance with the "state of metastatic grace", with particular attention to (i) reviewing how new imaging modalities impact the frontiers of conservative management, (ii) dissecting retinoblastoma genesis, growth patterns, and intraocular routes of tumor propagation, (iii) assessing major therapeutic changes and trends, (iv) proposing a classification of relapsing retinoblastoma, (v) examining treatable/preventable disease-related or treatment-induced complications, and (vi) appraising new therapeutic targets and concepts, as well as liquid biopsy potentiality.
Collapse
Affiliation(s)
- Francis L Munier
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland.
| | - Maja Beck-Popovic
- Unit of Pediatric Hematology-Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Guillermo L Chantada
- Hemato-Oncology Service, Hospital JP Garrahan, Buenos Aires, Argentina; Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain; Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - David Cobrinik
- The Vision Center and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; USC Roski Eye Institute, Department of Biochemistry & Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Tero T Kivelä
- Department of Ophthalmology, Ocular Oncology and Pediatric Ophthalmology Services, Helsinki University Hospital, Helsinki, Finland
| | - Dietmar Lohmann
- Eye Oncogenetics Research Group, Institute of Human Genetics, University Hospital Essen, Essen, Germany
| | - Philippe Maeder
- Unit of Neuroradiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Annette C Moll
- UMC, Vrije Universiteit Amsterdam, Department of Ophthalmology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Angel Montero Carcaboso
- Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain; Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Alexandre Moulin
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Paula Schaiquevich
- Unit of Clinical Pharmacokinetics, Hospital de Pediatria JP Garrahan, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Ciara Bergin
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Susan Houghton
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Francesco Puccinelli
- Interventional Neuroradiology Unit, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Yvan Vial
- Materno-Fetal Medicine Unit, Woman-Mother-Child Department, University Hospital of Lausanne, Switzerland
| | - Marie-Claire Gaillard
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Christina Stathopoulos
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
36
|
Rajasekaran S, Nagarajha Selvan LD, Dotts K, Kumar R, Rishi P, Khetan V, Bisht M, Sivaraman K, Krishnakumar S, Sahoo D, Campbell MJ, Elchuri SV, Miles WO. Non-coding and Coding Transcriptional Profiles Are Significantly Altered in Pediatric Retinoblastoma Tumors. Front Oncol 2019; 9:221. [PMID: 31058073 PMCID: PMC6477087 DOI: 10.3389/fonc.2019.00221] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Retinoblastoma is a rare pediatric tumor of the retina, caused by the homozygous loss of the Retinoblastoma 1 (RB1) tumor suppressor gene. Previous microarray studies have identified changes in the expression profiles of coding genes; however, our understanding of how non-coding genes change in this tumor is absent. This is an important area of research, as in many adult malignancies, non-coding genes including LNC-RNAs are used as biomarkers to predict outcome and/or relapse. To establish a complete and in-depth RNA profile, of both coding and non-coding genes, in Retinoblastoma tumors, we conducted RNA-seq from a cohort of tumors and normal retina controls. This analysis identified widespread transcriptional changes in the levels of both coding and non-coding genes. Unexpectedly, we also found rare RNA fusion products resulting from genomic alterations, specific to Retinoblastoma tumor samples. We then determined whether these gene expression changes, of both coding and non-coding genes, were also found in a completely independent Retinoblastoma cohort. Using our dataset, we then profiled the potential effects of deregulated LNC-RNAs on the expression of neighboring genes, the entire genome, and on mRNAs that contain a putative area of homology. This analysis showed that most deregulated LNC-RNAs do not act locally to change the transcriptional environment, but potentially function to modulate genes at distant sites. From this analysis, we selected a strongly down-regulated LNC-RNA in Retinoblastoma, DRAIC, and found that restoring DRAIC RNA levels significantly slowed the growth of the Y79 Retinoblastoma cell line. Collectively, our work has generated the first non-coding RNA profile of Retinoblastoma tumors and has found that these tumors show widespread transcriptional deregulation.
Collapse
Affiliation(s)
- Swetha Rajasekaran
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States.,The Ohio State University Comprehensive Cancer Center, Columbus, OH,, United States.,Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| | | | - Kathleen Dotts
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States.,The Ohio State University Comprehensive Cancer Center, Columbus, OH,, United States.,Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| | - Ranjith Kumar
- Department of Nanobiotechnology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Pukhraj Rishi
- Shri Bhagwan Mahavir Vitreoretinal Services and Ocular Oncology Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Vikas Khetan
- Shri Bhagwan Mahavir Vitreoretinal Services and Ocular Oncology Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Madhoolika Bisht
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States.,The Ohio State University Comprehensive Cancer Center, Columbus, OH,, United States.,Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| | | | | | - Debashis Sahoo
- Department of Pediatrics and Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA, United States
| | - Moray J Campbell
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Sailaja V Elchuri
- Department of Nanobiotechnology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Wayne O Miles
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States.,The Ohio State University Comprehensive Cancer Center, Columbus, OH,, United States.,Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
37
|
Athavale V, Khetan V. Knudson to embryo selection: A story of the genetics of retinoblastoma. Taiwan J Ophthalmol 2019; 8:196-204. [PMID: 30637191 PMCID: PMC6302566 DOI: 10.4103/tjo.tjo_37_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Retinoblastoma, the most common primary intraocular malignancy of the young, is a prototype hereditary cancer. Due to its fairly predictable Mendelian inheritance, easily examinable tumors, and early age of presentation, RB has served as the most extensively studied model for genetics in cancer. The genetic strides in RB have progressed at an exponential rate since the 1970s. The highly morbid, reasonably curable and distinctly predictable inheritance pattern of RB; not to mention its propensity to affect our young, forms the basis of aggressively encouraging genetic diagnosis in all patients of RB. In this article, we present the basic and clinically relevant concepts of the genetics retinoblastoma.
Collapse
Affiliation(s)
- Vrushali Athavale
- Department of Vitreoretina, Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Vikas Khetan
- Department of Vitreoretina, Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya, Chennai, Tamil Nadu, India
| |
Collapse
|
38
|
Dimaras H, Corson TW. Retinoblastoma, the visible CNS tumor: A review. J Neurosci Res 2019; 97:29-44. [PMID: 29314142 PMCID: PMC6034991 DOI: 10.1002/jnr.24213] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/02/2017] [Accepted: 12/11/2017] [Indexed: 12/11/2022]
Abstract
The pediatric ocular cancer retinoblastoma is the only central nervous system (CNS) tumor readily observed without specialized equipment: it can be seen by, and in, the naked eye. This accessibility enables unique imaging modalities. Here, we review this cancer for a neuroscience audience, highlighting these clinical and research imaging options, including fundus imaging, optical coherence tomography, ultrasound, and magnetic resonance imaging. We also discuss the subtype of retinoblastoma driven by the MYCN oncogene more commonly associated with neuroblastoma, and consider trilateral retinoblastoma, in which an intracranial tumor arises along with ocular tumors in patients with germline RB1 gene mutations. Retinoblastoma research and clinical care can offer insights applicable to CNS malignancies, and also benefit from approaches developed elsewhere in the CNS.
Collapse
Affiliation(s)
- Helen Dimaras
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Division of Clinical Public Health, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Child Health Evaluative Sciences Program, SickKids Research Institute, Toronto, ON, M5G 1X8, Canada
- Department of Human Pathology, College of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Timothy W. Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, 46202, USA
| |
Collapse
|
39
|
Gao R, Mitra RN, Zheng M, Wang K, Dahringer JC, Han Z. Developing Nanoceria-Based pH-Dependent Cancer-Directed Drug Delivery System for Retinoblastoma. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1806248. [PMID: 32699541 PMCID: PMC7375362 DOI: 10.1002/adfm.201806248] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Indexed: 05/17/2023]
Abstract
Development of a single combinatorial nano-platform technology to target cancer cells has been an unprecedented reality in boosting synergistic anti-tumor activities and in reducing off-target effects. We have designed a novel anti-tumor delivery system using a chemotherapy drug and a tumor target molecule covalently linked to cerium oxide nanoparticles (nanoceria). Nanoceria have a unique redox activity in that they possess antioxidant activity at physiological pH but have an intrinsic oxidase activity at acidic pH. Our system is integrated with (1) extracellular pH responsive functionality, (2) tumor cell targetable (CXC chemokine receptor 4, CXCR4 receptor specific) antagonist, (3) reactive oxygen species (ROS) inducible nanoceria, and (4) chemotherapeutic doxorubicin (DOX). These combinatorial nanoparticles (AMD-GCCNPs-DOX) are not only sensitive to the extracellular acidic pH conditions and targeted tumor cells but can also instantaneously induce ROS and release DOX intracellularly to enhance the chemotherapeutic activity in retinoblastoma cells (WERI-Rb-1 and Y79) and in xenograft (Y79/GFP-luc grafted) and genetic p107s (Rb Lox/lox , p107 +/- , p130 -/- ) orthotopic mice models. Together we introduce a lucidly engineered combinatorial nano-construct that offers a viable and simple strategy for delivering a cocktail of therapeutics into tumor cells under acidosis, exhibiting a promising new future for clinical therapeutic opportunities.
Collapse
Affiliation(s)
- Ruijuan Gao
- Department of Ophthalmology, University of North Carolina, 2208 Marsico Hall, 125 Mason Farm Rd, Chapel Hill, NC, USA 27599
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing, China 100050
| | - Rajendra Narayan Mitra
- Department of Ophthalmology, University of North Carolina, 2208 Marsico Hall, 125 Mason Farm Rd, Chapel Hill, NC, USA 27599
| | - Min Zheng
- Department of Ophthalmology, University of North Carolina, 2208 Marsico Hall, 125 Mason Farm Rd, Chapel Hill, NC, USA 27599
| | - Kai Wang
- Department of Ophthalmology, University of North Carolina, 2208 Marsico Hall, 125 Mason Farm Rd, Chapel Hill, NC, USA 27599
| | - Jesse Christine Dahringer
- Department of Ophthalmology, University of North Carolina, 2208 Marsico Hall, 125 Mason Farm Rd, Chapel Hill, NC, USA 27599
| | - Zongchao Han
- Department of Ophthalmology, University of North Carolina, 2208 Marsico Hall, 125 Mason Farm Rd, Chapel Hill, NC, USA 27599
- Carolina Institute for NanoMedicine, University of North Carolina, Chapel Hill, NC, USA 27599
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA 27599
| |
Collapse
|
40
|
Comprehensive characterization of RB1 mutant and MYCN amplified retinoblastoma cell lines. Exp Cell Res 2018; 375:92-99. [PMID: 30584916 DOI: 10.1016/j.yexcr.2018.12.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/19/2018] [Accepted: 12/21/2018] [Indexed: 12/26/2022]
Abstract
In retinoblastoma research tumor-derived cell lines remain an important model to investigate tumorigenesis and new therapy options, due to limited tumor material and lack of adequate animal models. A panel of 10 retinoblastoma cell lines was characterized with respect to mutation, methylation and expression of RB1 and MYCN. These established retinoblastoma cell lines represent the most frequent types of RB1 inactivation and together with the MYCN amplification status, three classes can be distinguished: RB1mut/MYCNnonA, RB1mut/MYCNA and RB1wt/MYCNA. MYCN amplification was identified in five cell lines, whereby two of them, RB522 and RB3823, harbor no aberration in RB1. Targeted sequencing of 160 genes often mutated in cancer identified only few variants in tumor-associated genes other than in RB1. None of these variants was recurrent. mRNA expression analyses of retinal markers, cell cycle regulators and members of the TP53 signaling pathway revealed a high variability between cell lines but no class-specific differences. The here presented thorough validation of retinoblastoma cell lines, including microsatellite analysis for cell line authentication, provides the basis for further in vitro studies on retinoblastoma.
Collapse
|
41
|
Saengwimol D, Rojanaporn D, Chaitankar V, Chittavanich P, Aroonroch R, Boontawon T, Thammachote W, Jinawath N, Hongeng S, Kaewkhaw R. A three-dimensional organoid model recapitulates tumorigenic aspects and drug responses of advanced human retinoblastoma. Sci Rep 2018; 8:15664. [PMID: 30353124 PMCID: PMC6199308 DOI: 10.1038/s41598-018-34037-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Persistent or recurrent retinoblastoma (RB) is associated with the presence of vitreous or/and subretinal seeds in advanced RB and represents a major cause of therapeutic failure. This necessitates the development of novel therapies and thus requires a model of advanced RB for testing candidate therapeutics. To this aim, we established and characterized a three-dimensional, self-organizing organoid model derived from chemotherapy-naïve tumors. The responses of organoids to drugs were determined and compared to relate organoid model to advanced RB, in terms of drug sensitivities. We found that organoids had histological features resembling retinal tumors and seeds and retained DNA copy-number alterations as well as gene and protein expression of the parental tissue. Cone signal circuitry (M/L+ cells) and glial tumor microenvironment (GFAP+ cells) were primarily present in organoids. Topotecan alone or the combined drug regimen of topotecan and melphalan effectively targeted proliferative tumor cones (RXRγ+ Ki67+) in organoids after 24-h drug exposure, blocking mitotic entry. In contrast, methotrexate showed the least efficacy against tumor cells. The drug responses of organoids were consistent with those of tumor cells in advanced disease. Patient-derived organoids enable the creation of a faithful model to use in examining novel therapeutics for RB.
Collapse
Affiliation(s)
- Duangporn Saengwimol
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duangnate Rojanaporn
- Department of Ophthalmology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Vijender Chaitankar
- Bioinformatics Computational Biology Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, USA
| | - Pamorn Chittavanich
- Section for Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rangsima Aroonroch
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tatpong Boontawon
- Section for Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Weerin Thammachote
- Section for Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Natini Jinawath
- Section for Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rossukon Kaewkhaw
- Section for Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
42
|
Goldsmith ZK, Coppess W, Irvine AS, Yuan K, Barsh SR, Ritter MK, McEwen MW, Flores-Otero J, Garcia-Vargas A, Martinez-Ferrer M, Brennan RC, Morales-Tirado VM, Wilson MW. Targeting the Platelet-Derived Growth Factor-beta Stimulatory Circuitry to Control Retinoblastoma Seeds. Invest Ophthalmol Vis Sci 2018; 59:4486-4495. [PMID: 30193324 PMCID: PMC6133233 DOI: 10.1167/iovs.18-24359] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022] Open
Abstract
Purpose Vitreous seeding remains the primary reason for treatment failure in eyes with retinoblastoma (Rb). Systemic and intra-arterial chemotherapy, each with its own inherent set of complications, have improved salvage rates for eyes with advanced disease, but the location and biology of vitreous seeds present a fundamental challenge in developing treatments with minimal toxicity and risk. The aim of this study was to target the platelet-derived growth factor (PDGF)- PDGF-receptor β (PDGFRβ) signaling pathway and investigate its role in the growth of Rb seeds, apoptotic activity, and invasive potential. Methods We performed ex vivo analyses on vitreous samples from Rb patients that underwent enucleation and from patient-derived xenografts. These samples were evaluated by quantitative PCR, immunohistochemistry, and ELISA. The effects of disruption of the PDGF-PDGFRβ signaling pathway, both by pharmacologic and genomic knockdown approaches, were evaluated in vitro by cell proliferation and apoptotic assays, quantitative PCR analyses, Western blotting, flow cytometry, and imaging flow cytometry. A three-dimensional cell culture system was generated for in-depth study of Rb seeds. Results Our results demonstrated that PDGFRβ signaling is active in the vitreous of Rb patients and patient-derived xenografts, sustaining growth and survival in an AKT-, MDM2-, and NF-κB-dependent manner. The novel three-dimensional cell culture system mimics Rb seeds, as the in vitro generated spheroids have similar morphologic features to Rb seeds and mimicked their natural physiology. Conclusions Targeting the PDGFRβ pathway in vitro reduces Rb cell growth, survival, and invasiveness and could augment current therapies. This represents a novel signaling pathway for potential targeted therapy to further improve ocular survival in advanced Rb.
Collapse
Affiliation(s)
- Zachary K. Goldsmith
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - William Coppess
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Andrew S. Irvine
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Kelley Yuan
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Samuel R. Barsh
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Madison K. Ritter
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Biology, Furman University, Greenville, South Carolina, United States
| | - Matthew W. McEwen
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Jacqueline Flores-Otero
- Department of Anatomy and Neurobiology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico, United States
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico, United States
| | - Aileen Garcia-Vargas
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico, United States
- Department of Pharmacology and Toxicology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico, United States
| | - Magaly Martinez-Ferrer
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico, United States
- Department of Pharmaceutical Sciences, University of Puerto Rico School of Pharmacy, San Juan, Puerto Rico, United States
| | - Rachel C. Brennan
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Vanessa M. Morales-Tirado
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Matthew W. Wilson
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| |
Collapse
|
43
|
Ge Y, Fuchs E. Stretching the limits: from homeostasis to stem cell plasticity in wound healing and cancer. Nat Rev Genet 2018; 19:311-325. [PMID: 29479084 PMCID: PMC6301069 DOI: 10.1038/nrg.2018.9] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cells (SCs) govern tissue homeostasis and wound repair. They reside within niches, the special microenvironments within tissues that control SC lineage outputs. Upon injury or stress, new signals emanating from damaged tissue can divert nearby cells into adopting behaviours that are not part of their homeostatic repertoire. This behaviour, known as SC plasticity, typically resolves as wounds heal. However, in cancer, it can endure. Recent studies have yielded insights into the orchestrators of maintenance and lineage commitment for SCs belonging to three mammalian tissues: the haematopoietic system, the skin epithelium and the intestinal epithelium. We delineate the multifactorial determinants and general principles underlying the remarkable facets of SC plasticity, which lend promise for regenerative medicine and cancer therapeutics.
Collapse
Affiliation(s)
- Yejing Ge
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| |
Collapse
|
44
|
Xie C, Freeman MJ, Lu H, Wang X, Forster CL, Sarver AL, Hallstrom TC. Retinoblastoma cells activate the AKT pathway and are vulnerable to the PI3K/mTOR inhibitor NVP-BEZ235. Oncotarget 2018; 8:38084-38098. [PMID: 28445155 PMCID: PMC5503517 DOI: 10.18632/oncotarget.16970] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/10/2017] [Indexed: 12/14/2022] Open
Abstract
Retinoblastoma is a pediatric cancer of the retina most often caused by inactivation of the retinoblastoma (RB1) tumor suppressor gene. We previously showed that Rb1 loss cooperates with either co-activating the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, or co-deleting Pten, to initiate retinoblastoma tumors in mice. The objectives of this study were to determine if the AKT pathway is activated in human retinoblastomas and the extent that anti-PI3K therapy induces apoptosis in retinoblastoma cells, alone or in combination with the DNA damaging drugs carboplatin and topotecan. Serial sections from human retinoblastoma tissue microarrays containing 27 tumors were stained with antibodies specific to p-AKT, Ki-67, forkhead box O1 (p-FOXO1), and ribosomal protein S6 (p-S6) using immunohistochemistry and each tumor sample scored for intensity. Human retinoblastoma tumors displayed significant correlation between p-AKT intensity with highly proliferative tumors (p = 0.008) that were also highly positive for p-FOXO1 (p = 0.002). Treatment with BEZ235, a dual PI3K/mTOR inhibitor, reduced phosphorylation levels of the AKT targets p-FOXO and p-S6 and effectively induced apoptosis the Y79 and Weri-1 human retinoblastoma cell lines and in vivo in our retinoblastoma mouse model. Long-term treatment with BEZ235 in vivo using our retinoblastoma-bearing mice induced apoptosis but did not significantly extend the lifespan of the mice. We then co-administered BEZ235 with topotecan and carboplatin chemotherapeutics in vivo, which more effectively induced apoptosis of retinoblastoma, but not normal retinal cells than either treatment alone. Our study has increased the variety of potentially effective targeted treatments that can be considered for human retinoblastoma.
Collapse
Affiliation(s)
- Chencheng Xie
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Matthew J Freeman
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Huarui Lu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Xiaohong Wang
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Colleen L Forster
- BioNet, Academic Health Center, University of Minnesota, Minneapolis, MN, USA
| | - Aaron L Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
45
|
Kan G, He H, Zhao Q, Li X, Li M, Yang H, Kim JK. Functional dissection of the role of UHRF1 in the regulation of retinoblastoma methylome. Oncotarget 2018; 8:39497-39511. [PMID: 28467809 PMCID: PMC5503627 DOI: 10.18632/oncotarget.17078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
UHRF1 (ubiquitin-like with PHD and RING finger domains 1) is a critical regulator for DNA methylation, and its frequent overexpression in human cancers has been associated with tumor-promoting effects. However, whether the overexpressed UHRF1 contributes to the establishment and maintenance of tumor methylomes and whether this process can affect the tumorigenesis remain unclear. In this study, we show that UHRF1 is highly expressed in retinoblastoma, and genomes of human primary retinoblastoma and cell lines have differential DNA methylation patterns compared with those of normal retina, characterized by lower global methylation and higher promoter methylation of tumor suppressors. However, our genome-wide DNA methylation study uncovers that UHRF1 down-modulation in retinoblastoma cells exerts minor effects on the existing methylation patterns at both bulk genome and individual gene loci, suggesting that retinoblastoma methylome is primarily maintained by other mechanisms. Furthermore, using two murine retinoblastoma models, we found that high UHRF1 expression does not alter global methylation levels in both premalignant neonatal retina and retinoblastoma tumors, implying that DNA hypomethylation may not be an early mechanism driving retinoblastoma tumorigenesis unlike what has been proposed for other types of cancer. These results suggest that tumor-promoting functions of UHRF1 in retinoblastoma are largely independent of its role in DNA methylation.
Collapse
Affiliation(s)
- Guangyan Kan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510060, China
| | - Heng He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Qi Zhao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Xiubo Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Min Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Huasheng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Jong Kyong Kim
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| |
Collapse
|
46
|
Garancher A, Lin CY, Morabito M, Richer W, Rocques N, Larcher M, Bihannic L, Smith K, Miquel C, Leboucher S, Herath NI, Dupuy F, Varlet P, Haberler C, Walczak C, El Tayara N, Volk A, Puget S, Doz F, Delattre O, Druillennec S, Ayrault O, Wechsler-Reya RJ, Eychène A, Bourdeaut F, Northcott PA, Pouponnot C. NRL and CRX Define Photoreceptor Identity and Reveal Subgroup-Specific Dependencies in Medulloblastoma. Cancer Cell 2018; 33. [PMID: 29533784 PMCID: PMC6368680 DOI: 10.1016/j.ccell.2018.02.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer cells often express differentiation programs unrelated to their tissue of origin, although the contribution of these aberrant phenotypes to malignancy is poorly understood. An aggressive subgroup of medulloblastoma, a malignant pediatric brain tumor of the cerebellum, expresses a photoreceptor differentiation program normally expressed in the retina. We establish that two photoreceptor-specific transcription factors, NRL and CRX, are master regulators of this program and are required for tumor maintenance in this subgroup. Beyond photoreceptor lineage genes, we identify BCL-XL as a key transcriptional target of NRL and provide evidence substantiating anti-BCL therapy as a rational treatment opportunity for select MB patients. Our results highlight the utility of studying aberrant differentiation programs in cancer and their potential as selective therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Alexandra Garancher
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Charles Y Lin
- Baylor College of Medicine, Department of Molecular and Human Genetics, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Morgane Morabito
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Wilfrid Richer
- PSL Research University, Paris, France; Institut Curie, Paris 75248, France; INSERM U830, Paris 75248, France; Translational Research in Pediatric Oncology, Institut Curie SiRIC, Paris, France
| | - Nathalie Rocques
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Magalie Larcher
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Laure Bihannic
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France; Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS325, Memphis, TN 38017, USA
| | - Kyle Smith
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS325, Memphis, TN 38017, USA
| | - Catherine Miquel
- Departement de Neuropathologie, Hôpital Sainte-Anne, INSERM UMR_S1165, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sophie Leboucher
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France
| | - Nirmitha I Herath
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Fanny Dupuy
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Pascale Varlet
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departement de Neuropathologie, Hôpital Sainte-Anne, INSERM U894, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Christine Walczak
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France; INSERM U1196, CNRS UMR9187, Centre Universitaire, Orsay 91405, France
| | - Nadine El Tayara
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France; INSERM U1196, CNRS UMR9187, Centre Universitaire, Orsay 91405, France
| | - Andreas Volk
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France; INSERM U1196, CNRS UMR9187, Centre Universitaire, Orsay 91405, France
| | - Stéphanie Puget
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Necker-Enfants Malades, Département Neurochirurgie pédiatrique, Paris, France
| | - François Doz
- Institut Curie, Paris 75248, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; SIREDO Center (Care, innovation, Research in pediatric, adolescent and young adult oncology), Institut Curie, Paris, France
| | - Olivier Delattre
- PSL Research University, Paris, France; Institut Curie, Paris 75248, France; INSERM U830, Paris 75248, France; SIREDO Center (Care, innovation, Research in pediatric, adolescent and young adult oncology), Institut Curie, Paris, France
| | - Sabine Druillennec
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Olivier Ayrault
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Robert J Wechsler-Reya
- Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alain Eychène
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France
| | - Franck Bourdeaut
- PSL Research University, Paris, France; Institut Curie, Paris 75248, France; INSERM U830, Paris 75248, France; Translational Research in Pediatric Oncology, Institut Curie SiRIC, Paris, France; SIREDO Center (Care, innovation, Research in pediatric, adolescent and young adult oncology), Institut Curie, Paris, France
| | - Paul A Northcott
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS325, Memphis, TN 38017, USA.
| | - Celio Pouponnot
- Institut Curie - Recherche, Laboratoire 110, Centre Universitaire, Orsay Cedex 91405, France; INSERM U1021, Centre Universitaire, Orsay 91405, France; CNRS UMR 3347, Centre Universitaire, Orsay 91405, France; Université Paris Sud-11, 91405 Orsay, France; PSL Research University, Paris, France.
| |
Collapse
|
47
|
Chen S, Poyer F, Garcia G, Fiorini-Debuisschert C, Rosilio V, Maillard P. Amphiphilic Glycoconjugated Porphyrin Heterodimers as Two-Photon Excitable Photosensitizers: Design, Synthesis, Photophysical and Photobiological Studies. ChemistrySelect 2018. [DOI: 10.1002/slct.201703013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Su Chen
- Department Chemistry and Modelisation and Imaging for Biology (CMIB); Institut Curie, Research Center; PSL Research University, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
- CNRS UMR 9187 - INSERM U 1196; Université Paris-Saclay; Université Paris Sud 11, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
| | - Florent Poyer
- Department Chemistry and Modelisation and Imaging for Biology (CMIB); Institut Curie, Research Center; PSL Research University, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
- CNRS UMR 9187 - INSERM U 1196; Université Paris-Saclay; Université Paris Sud 11, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
| | - Guillaume Garcia
- Department Chemistry and Modelisation and Imaging for Biology (CMIB); Institut Curie, Research Center; PSL Research University, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
- CNRS UMR 9187 - INSERM U 1196; Université Paris-Saclay; Université Paris Sud 11, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
| | | | - Véronique Rosilio
- Institut Galien Paris Sud, CNRS UMR 8612; Université Paris Sud 11; Université Paris-Saclay; 5 rue J.-B. Clément F-92296 Châtenay-Malabry France
| | - Philippe Maillard
- Department Chemistry and Modelisation and Imaging for Biology (CMIB); Institut Curie, Research Center; PSL Research University, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
- CNRS UMR 9187 - INSERM U 1196; Université Paris-Saclay; Université Paris Sud 11, Bât 110-112, Centre Universitaire; Rue Henri Becquerel F-91405 Orsay Cedex France
| |
Collapse
|
48
|
Structural changes of the macula and optic nerve head in the remaining eyes after enucleation for retinoblastoma: an optical coherence tomography study. BMC Ophthalmol 2017; 17:251. [PMID: 29246122 PMCID: PMC5732405 DOI: 10.1186/s12886-017-0650-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/05/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND To describe objectively the possible structural changes of the macula and optic nerve head in the free eyes of unilateral cured retinoblastoma patients and, also after enucleation using spectral domain optical coherence tomography. METHODS A cross sectional study involving 60 patients subdivided into three groups; 15 unilateral RB patients in whom enucleation was indicated as a sole treatment performed earlier in life [(study group (I)], 15 unilateral RB patients who had completely regressed disease with a preserved eye [(study group (II)] and 30 age and sex matched healthy controls. The remaining and free eyes in study groups and right eyes of control group had full ophthalmological examination, static automated perimetry and optical coherence tomography of the macula and optic nerve head. RESULTS In study group (II); a significant thinning of total macula, central fovea, ganglion cell layer (GCL), ganglion cell complex (GCC), and some sectors of outer nuclear layer (P- values ≤0.05) was found with no significant difference in peripapillary nerve fiber layer (pRNFL) thickness and optic nerve head parameters compared to the control group and the study group (I). A significantly thickened total macula, GCL, GCC, and pRNFL in study group (I) compared to study group (II). Thickened pRNFL was significantly correlated to standard automated perimetry pattern deviations. No significant difference was found between study group (I) and control group. CONCLUSION Retinoblastoma eyes characterized by thinning of central fovea, GCL, GCC compared to the control group. After unilateral enucleation, increased GCC and pRNFL thicknesses were detected compared to retinoblastoma group.
Collapse
|
49
|
Stenfelt S, Blixt MKE, All-Ericsson C, Hallböök F, Boije H. Heterogeneity in retinoblastoma: a tale of molecules and models. Clin Transl Med 2017; 6:42. [PMID: 29124525 PMCID: PMC5680409 DOI: 10.1186/s40169-017-0173-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/26/2017] [Indexed: 12/13/2022] Open
Abstract
Retinoblastoma, an intraocular pediatric cancer, develops in the embryonic retina following biallelic loss of RB1. However, there is a wide range of genetic and epigenetic changes that can affect RB1 resulting in different clinical outcomes. In addition, other transformations, such as MYCN amplification, generate particularly aggressive tumors, which may or may not be RB1 independent. Recognizing the cellular characteristics required for tumor development, by identifying the elusive cell-of-origin for retinoblastoma, would help us understand the development of these tumors. In this review we summarize the heterogeneity reported in retinoblastoma on a molecular, cellular and tissue level. We also discuss the challenging heterogeneity in current retinoblastoma models and suggest future platforms that could contribute to improved understanding of tumor initiation, progression and metastasis in retinoblastoma, which may ultimately lead to more patient-specific treatments.
Collapse
Affiliation(s)
- Sonya Stenfelt
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden
| | - Maria K E Blixt
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden
| | | | - Finn Hallböök
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden
| | - Henrik Boije
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden.
| |
Collapse
|
50
|
Lonfat N, Cepko C. Epigenomics of Retinal Development in Mice and Humans. Neuron 2017; 94:420-423. [PMID: 28472646 DOI: 10.1016/j.neuron.2017.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In this issue of Neuron, Aldiri et al. (2017) present an analysis of epigenetic changes during retinal development, and use these data to probe reprogramming of retinal iPSC cells, as well as the origin of retinoblastoma cells.
Collapse
Affiliation(s)
- Nicolas Lonfat
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Connie Cepko
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|