human retinoblastoma
Recently Published Documents


TOTAL DOCUMENTS

306
(FIVE YEARS 43)

H-INDEX

38
(FIVE YEARS 3)

2022 ◽  
Author(s):  
Sumera Perveen ◽  
Carlos A Zepeda-Velazquez ◽  
David McLeod ◽  
Richard Marcellus ◽  
Mohammed Mohammed ◽  
...  

RBBP4 is a nuclear WD40 motif-containing protein widely implicated in various cancers and a putative drug target. It interacts with multiple proteins within diverse complexes such as nucleosome remodeling and deacetylase (NuRD) complex and polycomb repressive complex 2 (PRC2), as well as histone H3 and H4 through two distinct binding sites. B-cell lymphoma/leukemia 11A (BCL11A), friend of GATA-1 (FOG-1), plant homeodomain finger protein 6 (PHF6) and histone H3 bind to the top of the donut-shaped seven-bladed β-propeller fold of RBBP4, while suppressor of zeste 12 (SUZ12), metastasis associated protein 1 (MTA1) and histone H4 bind to a pocket on the side of the WD40 repeats of this protein. Here, we report the discovery of the first small molecule antagonists of the RBBP4 top pocket, competing with interacting peptides from proteins such as BCL11A and histone H3. We also determined the first crystal structure of RBBP4 in complex with a small molecule (OICR17251), paving the path for structure-guided design and optimization towards more potent antagonists.


2021 ◽  
Vol 12 (12) ◽  
Author(s):  
Jie Yang ◽  
Yongyun Li ◽  
Yanping Han ◽  
Yiyi Feng ◽  
Min Zhou ◽  
...  

AbstractRetinoblastoma is a childhood retinal tumour that is the most common primary malignant intraocular tumour. However, it has been challenging to identify the cell types associated with genetic complexity. Here, we performed single-cell RNA sequencing on 14,739 cells from two retinoblastoma samples to delineate the heterogeneity and the underlying mechanism of retinoblastoma progression. Using a multiresolution network-based analysis, we identified two major cell types in human retinoblastoma. Cell trajectory analysis yielded a total of 5 cell states organized into two main branches, and the cell cycle-associated cone precursors were the cells of origin of retinoblastoma that were required for initiating the differentiation and malignancy process of retinoblastoma. Tumour cells differentiation reprogramming trajectory analysis revealed that cell-type components of multiple tumour-related pathways and predominantly expressed UBE2C were associated with an activation state in the malignant progression of the tumour, providing a potential novel “switch gene” marker during early critical stages in human retinoblastoma development. Thus, our findings improve our current understanding of the mechanism of retinoblastoma progression and are potentially valuable in providing novel prognostic markers for retinoblastoma.


Author(s):  
Marco Zschoche ◽  
Sergej Skosyrski ◽  
Neele Babst ◽  
Mahdy Ranjbar ◽  
Felix Rommel ◽  
...  
Keyword(s):  

Cancers ◽  
2021 ◽  
Vol 13 (20) ◽  
pp. 5248
Author(s):  
Swatishree Sradhanjali ◽  
Padmalochan Rout ◽  
Devjyoti Tripathy ◽  
Swathi Kaliki ◽  
Suryasnata Rath ◽  
...  

Retinoblastoma is usually initiated by biallelic RB1 gene inactivation. In addition, MYCN copy number alterations also contribute to RB pathogenesis. However, MYCN expression, its role in disease progression and correlation with RB histological risk factors are not well understood. We studied the expression of MYCN in enucleated RB patient specimens by immunohistochemistry. MYCN is overexpressed in RB compared to control retina. Our microarray gene expression analysis followed by qRT-PCR validation revealed that genes involved in glucose metabolism and migration are significantly downregulated in MYCN knockdown cells. Further, targeting MYCN in RB cells using small molecule compounds or shRNAs led to decreased cell survival and migration, increased apoptosis and cell cycle arrest, suggesting that MYCN inhibition can be a potential therapeutic strategy. We also noted that MYCN inhibition results in reduction in glucose uptake, lactate production, ROS levels and gelatinolytic activity of active-MMP9, explaining a possible mechanism of MYCN in RB. Taking clues from our findings, we tested a combination treatment of RB cells with carboplatin and MYCN inhibitors to find enhanced therapeutic efficacy compared to single drug treatment. Thus, MYCN inhibition can be a potential therapeutic strategy in combination with existing chemotherapy drugs to restrict tumor cell growth in RB.


Author(s):  
Marco Zschoche ◽  
Sergej Skosyrski ◽  
Neele Babst ◽  
Mahdy Ranjbar ◽  
Felix Rommel ◽  
...  

Abstract Background The role of CD133 und ABCB5 is discussed in treatment resistance in several types of cancer. The objective of this study was to evaluate whether CD133+/ABCB5+ colocalization differs in untreated, in beam radiation treated, and in chemotherapy treated retinoblastoma specimens. Additionally, CD133, ABCB5, sphingosine kinase 1, and sphingosine kinase 2 gene expression was analyzed in WERI-RB1 (WERI RB1) and etoposide-resistant WERI RB1 subclones (WERI ETOR). Methods Active human untreated retinoblastoma specimens (n = 12), active human retinoblastoma specimens pretreated with beam radiation before enucleation (n = 8), and active human retinoblastoma specimens pretreated with chemotherapy before enucleation (n = 7) were investigated for localization and expression of CD133 and ABCB5 by immunohistochemistry. Only specimens with IIRC D, but not E, were included in this study. Furthermore, WERI RB1 and WERI ETOR cell lines were analyzed for CD133, ABCB5, sphingosine kinase 1, and sphingosine kinase 2 by the real-time polymerase chain reaction (RT-PCR). Results Immunohistochemical analysis revealed the same amount of CD133+/ABCB5+ colocalization islets in untreated and treated human retinoblastoma specimens. Quantitative RT-PCR analysis showed a statistically significant upregulation of CD133 in WERI ETOR (p = 0.002). No ABCB5 expression was detected in WERI RB1 and WERI ETOR. On the other hand, SPHK1 (p = 0.0027) and SPHK2 (p = 0.017) showed significant downregulation in WERI ETOR compared to WERI RB1. Conclusions CD133+/ABCB5+ co-localization islets were noted in untreated and treated human retinoblastoma specimens. Therefore, we assume that CD133+/ABCB5+ islets might play a role in retinoblastoma genesis, but not in retinoblastoma treatment resistance.


2021 ◽  
Vol 46 (5) ◽  
Author(s):  
Shu Liu ◽  
Chunmei Hu ◽  
Yingxue Wang ◽  
Guang Shi ◽  
Yarong Li ◽  
...  

2021 ◽  
Author(s):  
Na Zhao ◽  
Lei Zhou ◽  
Qinkang Lu ◽  
Shengzhan Wang ◽  
Yanli Sun ◽  
...  

Abstract Background: Cancer stem cells are responsible for tumor initiation and progression in various types of cancer, while, although the existence of retinoblastoma stem cells had been reported, the mechanisms supporting retinoblastoma stemness are poorly understood. In this study, a modified method for isolating retinoblastoma stem-like cells for mechanistic study was first established and an important mechanism underlying SOX2-drived retinoblastoma stemness was subsequently revealed.Methods: The retinoblastoma stem-like cells were isolated by single cell cloning in combination of examination of sphere-forming capacities. The stemness of isolated retinoblastoma stem-like cells were characterized by sphere-forming capacities and the expression of cancer stem cell markers. The SOX2 gene was overexpressed or knocked down by lentivirus system. The transcriptional regulation was identified by qRT-PCR, luciferase reporter, nuclear run-on and DNA pull down assay. Spearman analysis was employed for correlation analysis of genes in tumor tissues of retinoblastoma patients. Results: The isolated retinoblastoma stem-like cells exhibited significantly enhanced sphere-forming capacity and constantly higher levels of CD44, ABCG2, SOX2 and PAX6, but not CD133. SOX2 positively regulated the stemness of retinoblastoma stem-like cells as identified by gene manipulation technology. SOX2 directly binds to the promoters of WWTR1 and YAP1, transcriptionally activates WWTR1 and YAP1, and thereby activating Hippo/YAP signaling. Knockdown of WWTR1 or YAP1 partially abolished the effect of SOX2 on the stemness of retinoblastoma stem-like cells. Conclusion: An effective method for isolation of retinoblastoma stem-like cells was established. The mechanistic study demonstrated that SOX2, as a key deriver, maintains retinoblastoma stemness by activating Hippo/YAP signaling. Inhibition of Hippo/YAP signaling would be an effective strategy for human retinoblastoma caused by aberrant upregulation of SOX2.


2021 ◽  
Vol 17 (6) ◽  
pp. 420-427
Author(s):  
Weifang Cao ◽  
Feiyan Chai ◽  
Ling Tong ◽  
Xiaoyong Yuan

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Jackie L. Norrie ◽  
Anjana Nityanandam ◽  
Karen Lai ◽  
Xiang Chen ◽  
Matthew Wilson ◽  
...  

AbstractRetinoblastoma 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.


Sign in / Sign up

Export Citation Format

Share Document