B A Spengler's research while affiliated with Fordham University and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (94)


Cytogenetic Consequences of Gene Amplification
  • Chapter

August 2020

·

12 Reads

June L. Biedler

·

Barbara A. Spengler
Share

Reciprocal antagonistic regulation of N-myc mRNA by miR‑17 and the neuronal-specific RNA-binding protein HuD

May 2017

·

7 Reads

·

10 Citations

Oncology Reports

Neuroblastoma is a childhood cancer originating from embryonic neural crest cells. Amplification of the proto‑oncogene N-myc, seen in ~30% of neuroblastoma tumors, is a marker for poor prognosis. Recently discovered small regulatory RNAs, microRNAs (miRNAs), are implicated in cancers, including neuroblastoma. miRNAs downregulate the expression of genes by binding to the 3'-untranslated regions (3'-UTRs), thereby inhibiting translation or inducing degradation of cognate mRNAs. Our study sought to identify miRNAs that regulate N-myc expression and thereby malignancy in neuroblastoma. miRNAs whose expression negatively correlates with N-myc expression were identified from a miRNA microarray of 4 N-myc-amplified neuroblastoma cell lines. Three of these miRNAs (miR-17, miR-20a and miR-18a) belong to the miR-17-92 cluster, previously shown to be upregulated by N-myc. qPCR validation of these miRNAs in a larger panel of cell lines revealed that levels of miR-17 were inversely proportional to N-myc mRNA amounts in the N-myc-amplified cell lines. Notably, miR-17 also downregulated N-myc protein synthesis in the N-myc-amplified cells, thereby generating a negative feedback regulatory loop between the proto-oncogene and this miRNA. Moreover, the neuronal-specific RNA-binding protein HuD (ELAVL4), which regulates the processing/stability of N-myc mRNA, competes with miR-17 for a binding site in the 3'-UTR of N-myc. Thus, N-myc levels appear to be modulated by the antagonistic interactions of both miR-17, as a negative regulator, and HuD, as a positive regulator, providing further evidence of the complex cellular control mechanisms of this oncogene in N-myc-amplified neuroblastoma cells.


Unsupervised clustering based on miRNA expression profiles was generated using MultiExperiment Viewer (MeV) version 4 (http://www.tm4.org/mev.html) and shows that N- and I-type cells are distinctly different from miRNA mix containing S-type cells.
qRT-PCR analysis of phenotype-specific miRNAs in NB cell lines. The cell line panel includes six N-type [SH-SY5Y (1), SMS-LHN (2), BE(2)-M17V (3), LA1-55n (4), KCN-83n (5), SK-N-BE(1)n (6)]; four I-type [CB-JMN (7), BE(2)-C (8), SK-N-LD (9), SK-N-HM (10)]; and three S-type cell lines [SH-EP1 (11), SMS-KCNs (12), and LA1-5s (13)]. Levels of S-cell specific (A) miR-21, (B) -221 and (C) -335 were normalized to loading control U6 and expressed as a fold change compared to a standard sample of LA1-5s. Levels of (D) miR-124 and (E) -375 were normalized to loading control U6 and expressed as a fold change compared to a SH-SY5Y standard. Each bar represents the mean ± SEM of 3 or more samples.
Drug-induced irreversible differentiation of I-type NB cancer stem cell confirms the association miRNAs with cell phenotype. A. Fold increases in miR-21, -221 and -335 in BE(2)-C cells differentiated to an S phenotype following a 2 week treatment with 10-5 M BrdU. B. Changes in miR-124 and -375 expression in BE(2)-C cells treated with RA or BrdU to induce an N or S phenotype, respectively. Each bar represents the mean ± SEM of 4–6 determinations normalized to untreated controls set =1.0.
MiR-335 regulates expression of HAND1 and JAG1- modulators of neuronal differentiation. A. Quantitative changes in miR-335, HAND1, and JAG1 expression in miR-335 inhibitor-treated SH-EP1 cells. Each bar represents the mean ± SEM of three independent experiments. B. Semi-quantitative RT-PCR analysis of mRNA expression of HAND1 (B) and JAG1 (C) in cell lines of different phenotypes.
miR-124 induces neuronal differentiation in I-type NB cells. A. The miR-124 levels in control and miR-124-infected BE(2)-C. B. Immunofluorescence microscopy of BE(2)-C cells infected with a lentiviral-vector expressing GFP (control) or (C) BE(2)-C cells infected with lentiviral vector co-expressing miR-124 and GFP. Photomicrographs were taken two weeks after infection. Note the increase in number and size of multiple neuritic processes in C (arrows). D. 3H-NE uptake in BE(2)-C cells is increased 1.5–fold (P < 0.002) following treatment with RA and 3.7-fold (P < 0.001) with miR-124 lentiviral infection. E. N-myc mRNA levels are decreased ~2-fold (P < 0.008) in BE(2)-C/miR-124 lentiviral vector cells compared to control. F. Colony forming efficiencies (CFE) of BE(2)-C cells stably infected with miR-124 lentiviral vector or control. Note that CFE is reduced nearly 6-fold following infection (P < 0.001).

+2

MicroRNAs define distinct human neuroblastoma cell phenotypes and regulate their differentiation and tumorigenicity
  • Article
  • Full-text available

May 2014

·

98 Reads

·

38 Citations

BMC Cancer

Leleesha Samaraweera

·

Kathryn B Grandinetti

·

Ruojun Huang

·

[...]

·

Robert A Ross

Background Neuroblastoma (NB) is the most common extracranial solid tumor in children. NB tumors and derived cell lines are phenotypically heterogeneous. Cell lines are classified by phenotype, each having distinct differentiation and tumorigenic properties. The neuroblastic phenotype is tumorigenic, has neuronal features and includes stem cells (I-cells) and neuronal cells (N-cells). The non-neuronal phenotype (S-cell) comprises cells that are non-tumorigenic with features of glial/smooth muscle precursor cells. This study identified miRNAs associated with each distinct cell phenotypes and investigated their role in regulating associated differentiation and tumorigenic properties. Methods A miRNA microarray was performed on the three cell phenotypes and expression verified by qRT-PCR. miRNAs specific for certain cell phenotypes were modulated using miRNA inhibitors or stable transfection. Neuronal differentiation was induced by RA; non-neuronal differentiation by BrdU. Changes in tumorigenicity were assayed by soft agar colony forming ability. N-myc binding to miR-375 promoter was assayed by chromatin-immunoprecipitation. Results Unsupervised hierarchical clustering of miRNA microarray data segregated neuroblastic and non-neuronal cell lines and showed that specific miRNAs define each phenotype. qRT-PCR validation confirmed that increased levels of miR-21, miR-221 and miR-335 are associated with the non-neuronal phenotype, whereas increased levels of miR-124 and miR-375 are exclusive to neuroblastic cells. Downregulation of miR-335 in non-neuronal cells modulates expression levels of HAND1 and JAG1, known modulators of neuronal differentiation. Overexpression of miR-124 in stem cells induces terminal neuronal differentiation with reduced malignancy. Expression of miR-375 is exclusive for N-myc-expressing neuroblastic cells and is regulated by N-myc. Moreover, miR-375 downregulates expression of the neuronal-specific RNA binding protein HuD. Conclusions Thus, miRNAs define distinct NB cell phenotypes. Increased levels of miR-21, miR-221 and miR-335 characterize the non-neuronal, non-malignant phenotype and miR-335 maintains the non-neuronal features possibly by blocking neuronal differentiation. miR-124 induces terminal neuronal differentiation with reduction in malignancy. Data suggest N-myc inhibits neuronal differentiation of neuroblastic cells possibly by upregulating miR-375 which, in turn, suppresses HuD. As tumor differentiation state is highly predictive of patient survival, the involvement of these miRNAs with NB differentiation and tumorigenic state could be exploited in the development of novel therapeutic strategies for this enigmatic childhood cancer.

Download

Figure 1. Ras isoforms expressed in neuroblastoma cell variants. (A) Representative Western blot of total Ras protein in neuroblastoma cells using a pan-Ras antibody. Cell lines are N-type: 1) SH-SY5Y, 2) LA1-55n, 3) SK-N-BE(1)n; I-type: 4) BE(2)-C, 5) SK-N-HM, 6) SK-N-MM, 7) CB-JMN; and S-type: 8) SH-EP1, 9) LA1-5s, 10) SMS-KCNs. Actin is used as the loading control. (B) Representative Western blot of H/N-Ras, K-Ras, and actin in neuroblastoma cell lines of different phenotypes. Cell lines are N-type: 1) SH-SY5Y, 2) LA1-55n; I-type: 3) BE(2)-C, 4) SK-N-LD, 5) CB-JMN, 6) SK-N-MM; S-type: 7) SHEP1, 8) LA1-5s; and 9) control K-Ras-transfected LA1-5s. (C) Semiquantitative RT-PCR shows that N-Ras mRNA levels are much higher than H-Ras. DNA from plasmids containing N-or H-Ras coding sequence is included as controls (lane 12) to show a similar efficiency of primers. The cell lines are N-type: 1) SH-SY5Y, 2) LA1-55n, 3) SK-N-BE(1)n, 4) BE(2)-M17V; I-type: 5) SK-N-LD, 6) BE(2)-C, 7) SK-N-HM, 8) CB-JMN; and S-type: 9) SH-EP1, 10) LA1-5s, 11) SMS-KCNs. GAPD is used as the loading control. All lanes are from the same gel. (D) Four cell lines, SH-SY5Y (N-type), BE(2)-C (I-type), SK-N-LD (I-type), and LA1-5s (S-type), were chosen for quantitative real-time RT-PCR analysis. Serially diluted H-Ras and N-Ras plasmid constructs were used to generate standard curves. In all 4 cell lines, N-Ras mRNA levels are significantly 10-to 40-fold higher than H-Ras (**P < 0.001). (E) Representative Western blot of N-Ras, H-Ras, and actin in neuroblastoma cell lines of different phenotypes. Cell lines are the same as in B. Note that H-Ras mRNA and protein levels are much lower than those of N-Ras in all of the cell lines. 
Figure 2. Activated Ras-GTP levels are high in I-type neuroblastoma cells. (A) Representative Western blot of N-Ras-GTP levels in neuroblastoma cells. Cell lines are N-type: 1) SH-SY5Y, 2) LA1-55n, 3) SK-N-BE(1)n, 4) IMR-32; I-type: 5) BE(2)-C, 6) SK-N-LD, 7) SK-N-MM, 8) CB-JMN, 9) SKN-HM; and S-type: 10) SH-EP1, 11) LA1-5s, 12) SMS-KCNs. (B) Ras-GTP levels are significantly higher in I-type cells. n = number of different cell lines (*P < 0.01). Each cell line was examined in at least 3 independent experiments with different protein lysates and Ras-GTP levels normalized to total ras protein. (C) Representative Western blot shows that Ras-GTP levels decrease significantly in both RA-and BrdU-induced differentiated BE(2)-C cells (2.3-and 3.8-fold, respectively; P < 0.01). 
Figure 3. Changing Ras-GTP level by dominant-negative (DN) and constitutively active (CA) N-Ras constructs alters malignancy in neuroblastoma cells. DN-N-Ras-transfected I-type BE(2)-C clones have a (A) >2-fold decrease in Ras-GTP level and (B) 3.8-to 5.0-fold decrease in CFE in soft agar compared to the vector-transfected control (*P < 0.01). (C) Representative photomicrographs of the DN-N-Ras-or control vector-transfected BE(2)-C cells from the soft agar assay. Conversely, CA-N-Ras expression in N-type SH-SY5Y cells causes a (D) dramatic increase in Ras-GTP level (>10-fold) and (E) significant increase in CFE in soft agar (7.5-fold; **P < 0.001). (F) Representative photomicrographs of the CA-N-Ras-or control vector-transfected SH-SY5Y cells from the soft agar assay. 
Figure 4. High levels of Ras-GTP protect neuroblastoma cells from apoptosis. (A) Cell death was investigated using an apoptosis and necrosis assay, in which apoptotic cells exhibit green fluorescence, necrotic cells show red fluorescence, late-phase apoptotic cells show both green and red fluorescence, and living cells are not fluorescent. (Left) Representative phase-contrast photomicrograph used to determine the total cell number. (Right) A fluorescent photomicrograph of the same field showing the much higher rate of apoptosis in the DN-N-Ras-transfected BE(2)-C cells compared to the vector control. (B) In each experiment, apoptotic and necrotic cells were counted in 10 randomly chosen fields and expressed as a percentage of the total cell number. DN-N-Ras-transfected BE(2)-C cells show a significant, 4.1-fold, increase in apoptosis rate (*P < 0.01) but no change in necrosis. (C) Representative phase-contrast photomicrograph (left) and its fluorescent photomicrograph (right) showing the much lower rate of cell death in the CA-N-Ras-transfected SH-SY5Y cells compared to the vector control. (D) Percentage of apoptotic cells in CA-N-Ras-transfected SH-SY5Y cells is significantly, 4.0-fold, lower than the control (*P < 0.02), with no change in necrosis. 
Figure 7. Down-regulation of neurofibromin in I-type cells occurs at the posttranslational level. (A) Semiquantitative RT-PCR analysis using primers that recognize exons 58 and 59 of NF1 mRNA, coding for the epitope specifically recognized by the neurofibromin antibody, shows similar levels of NF1 mRNA in all cell variants. The cell lines are N-type: 1) SH-SY5Y, 2) LA1-55n, 3) IMR-32, 4) KCN-83n; I-type: 5) SK-N-LD, 6) BE(2)-C, 7) CBJMN, 8) SK-N-MM; and S-type: 9) SH-EP1, 10) LA1-5s, 11) SMS-KCNs. All lanes are from the same gel. (B) Real-time RT-PCR analyses using primers that amplify the N-terminal region (exon 2) of NF1 mRNA show that NF1 mRNA levels do not differ significantly among phenotypes. Relative NF1 mRNA level was normalized to GAPD mRNA and compared to SH-SY5Y set = 1.0. Each cell line was examined in 3 to 6 independent experiments with different RNA isolations (NS = not significantly different from N cell level; n = number of different cell lines). (C) I-type SK-N-MM and N-type SH-SY5Y cells were treated with MG132 at 10 μM for 3 and 6 hours. DMSO-treated cells were used as controls. Neurofibromin protein was detected by Western blotting. Actin is the negative control; hsp72 is the positive control. MG132 treatment significantly increases the neurofibromin amount in SK-N-MM cells 2.0-fold after 3 hours (P < 0.01) and 3.2-fold after 6 hours (P < 0.01). The neurofibromin amounts of SHSY5Y cells are not significantly changed after MG132 treatment. 
Increased Wild-Type N-Ras Activation by Neurofibromin Down-Regulation Increases Human Neuroblastoma Stem Cell Malignancy

November 2011

·

278 Reads

·

14 Citations

Genes & Cancer

Cellular heterogeneity is a well-known feature of human neuroblastoma tumors and cell lines. Of the 3 phenotypes (N-, I-, and S-type) isolated and characterized, the I-type cancer stem cell of neuroblastoma is the most malignant. Here, we report that, although wild-type N-Ras protein is expressed at the same level in all 3 neuroblastoma cell phenotypes, activated N-Ras-GTP level is significantly higher in I-type cancer stem cells. When activated N-Ras levels were decreased by transfection of a dominant-negative N-Ras construct, the malignant potential of I-type cancer stem cells decreased significantly. Conversely, when weakly malignant N-type cells were transfected with a constitutively active N-Ras construct, activated N-Ras levels, and malignant potential, were significantly increased. Thus, high levels of N-Ras-GTP are required for the increased malignancy of I-type neuroblastoma cancer stem cells. Moreover, increased activation of N-Ras results from significant down-regulation of neurofibromin (NF1), an important RasGAP. This specific down-regulation is mediated by an ubiquitin-proteasome-dependent pathway. Thus, decreased expression of NF1 in I-type neuroblastoma cancer stem cells causes a high level of activated N-Ras that is, at least in part, responsible for their higher tumorigenic potential.


Abstract B76: MicroRNAs regulate cell phenotype and N-myc expression in human neuroblastoma cells

December 2009

·

1 Read

Cancer Research

Identification of miRNAs that regulate neuroblast growth and differentiation could be important in understanding the onset and progression of the childhood cancer neuroblastoma, and in developing new therapeutics to fight this often fatal disease. Neuroblastoma tumors and derived cell lines are heterogeneous, with three basic phenotypes ‐ neuroblastic (N), substrate‐adherent (S), and cancer stem cells (I). These types differ significantly in their morphology, biochemistry and tumorigenic potential. Five miRNAs differentially expressed with respect to cell phenotype were identified from a miRNA microarray, validated by RT‐PCR, and quantified by real‐time RT‐PCR taqman assays in 14 different cell lines (6 N‐, 5 I‐ and 3 S‐types). Out of these miRNAs, three are highly expressed in non‐tumorigenic S‐type cell lines compared to I‐ and N‐type cell lines: miR‐21 (10‐ fold, p<0.05), miR‐221 (18‐fold, p<0.05) and miR‐335 (136‐fold, p<0.05). Moreover, miRNA expression increased significantly with BrdU‐induced I‐ to S‐type differentiation of BE(2)‐C. By contrast, miR‐124a levels are 10‐fold higher in N‐type cells compared to both I‐ and S‐type cells and are elevated 2‐fold (p<0.05) following retinoic acid‐induced neuronal differentiation of I‐type BE(2)‐C. Transient transfection of miR‐124a into I‐cells induced a neuronal‐like phenotype with increased neurite growth, supporting its active role in neuronal differentiation. Another neuronal‐specific miRNA, miR‐375, expressed in N‐ and I‐ but not in S‐type cells, significantly decreased with induced differentiation from an I‐ to S‐type phenotype. A second factor affecting survival of neuroblastoma patients is overexpression/amplification of the protooncogene N‐myc. Thus, the microarray was sorted for miRNAs whose levels correlated directly or inversely with N‐myc gene copy number. Candidate miRNAs were quantified by real‐time RT‐PCR taqman assays in the same 14 cell lines (8 N‐myc amplified and 6 non‐amplified). Three members of the miR‐17‐92 cluster (miR‐17‐5p, miR‐18a and miR‐20a) are highly expressed in N‐myc‐amplified cell lines compared to non‐amplified cells (p<0.05). Moreover, siRNA‐induced reduction of N‐myc protein (2.5‐fold) down‐regulated expression levels of all three miRNAs by 70% (p<0.05), suggesting N‐myc up‐regulates the expression of the miR‐17‐92 cluster. A screen of potential targets of the miR‐17‐92 cluster members revealed, interestingly, that N‐myc mRNA itself is a predicted target of miR‐17‐5p. Down‐regulation of this miRNA (95%) by transient transfection with specific inhibitors increased N‐myc mRNA 2‐fold (p<0.05) and protein 1.5‐fold (p<0.05), confirming and extending results of Cloonan et al (2008). In N‐myc amplified cell lines, N‐myc mRNA levels negatively correlate with levels of miR‐17‐5p. Together these data reveal a possible negative feedback loop between N‐myc and miR‐17‐5p. In conclusion, miRNAs that are differentially expressed with respect to both phenotype and N‐myc amplification status may significantly contribute to the progression and aggressiveness of neuroblastoma. Citation Information: Cancer Res 2009;69(23 Suppl):B76.


Human neuroblastoma stem cells

July 2007

·

22 Reads

·

120 Citations

Seminars in Cancer Biology

Human neuroblastoma is an embryonic cancer of the neural crest. Cellular heterogeneity is a characteristic feature of both tumors and derived cell lines. Recent studies have revealed that both cell lines and tumors contain cancer stem cells. In culture, these cells are self-renewing, multipotent, and highly malignant; in tumors their frequency correlates with a worse prognosis. Their identification and characterization should now permit a targeted approach to more effective treatment of this often fatal childhood cancer.


Loss of one HuD allele on chromosome #1p selects for amplification of the N-myc proto-oncogene in human neuroblastoma cells

March 2006

·

20 Reads

·

18 Citations

Oncogene

In human neuroblastoma tumors, amplification of the N-myc proto-oncogene and loss of all or part of the short arm of chromosome #1 are both associated with a poor prognosis. Accruing evidence indicates that it is the absence of one allele of the HuD (ELAVL4) gene, encoding the neuronal-specific RNA-binding protein HuD and localized to 1p34, that is linked to amplification. In 12 human neuroblastoma cell lines, N-myc amplification correlates with loss of one HuD allele and decreased HuD expression. Transfection experiments demonstrate that modulating HuD expression affects N-myc gene copy number as well as expression. Introduction of a sense HuD construct into two N-myc amplified cell lines considerably increases N-myc expression whereas gene copy number decreases. Conversely, expression of antisense HuD in N-myc nonamplified SH-SY5Y cells reduces HuD and N-myc mRNA levels even as cells show amplification of the N-myc gene. Thus, N-myc gene copy number is modulated by alteration of HuD expression. We propose that haploinsufficiency of HuD due to chromosome #1p deletion in neuroblastoma selects for cells that amplify N-myc genes. Application of these findings could lead to more effective therapies in the treatment of those patients with the worst prognosis.


Characteristics of Stem Cells from Human Neuroblastoma Cell Lines and in Tumors

November 2004

·

453 Reads

·

214 Citations

Neoplasia

Cellular heterogeneity is a hallmark of human neuroblastoma tumors and cell lines. Within a single neuroblastoma are cells from distinct neural crest lineages whose relative abundance is significant for prognosis. We postulate that a self-renewing multipotent tumor stem cell, which gives rise to diverse cell lineages, is the malignant progenitor of this cancer. To test this hypothesis, we have established 22 cloned, phenotypically homogeneous populations of the three major cell types from 17 neuroblastoma cell lines. In vitro, malignant neuroblastoma stem cells, termed I-type (intermediate type), have distinct morphologic, biochemical, differentiative, and tumorigenic properties. I-type cells express features of both neuroblastic (N) cells (scant cytoplasm, neuritic processes, neurofilaments, pseudoganglia, and granin and neurotransmitter enzyme expression) and substrate-adherent (S) cells (extensive cytoplasm and vimentin and CD44 expression). Moreover, they show bidirectional differentiation to either N or S cells when induced by specific agents. I-type cells are significantly more malignant than N- or S-type cells, with four- to five-fold greater plating efficiencies in soft agar and six-fold higher tumorigenicity in athymic mice. Differences in malignant potential are unrelated to N-myc amplification/overexpression or the ability to digest and migrate through the extracellular matrix. Immunocytochemical analyses of a small series of tumors reveal that frequency of cells coexpressing N and S cell markers correlates with poor prognosis. Thus, I-type stem cells may be instrumental in the genesis and growth of tumors in the patient. Their unique biology deserves attention and further investigation.


Alternative pathways of MYCN gene copy number increase in primary neuroblastoma tumors

September 2004

·

16 Reads

·

13 Citations

Cancer Genetics and Cytogenetics

Neuroblastomas, tumors of the sympathetic nervous system, account for 7-10% of the cancers of childhood. Genetic studies have shown, and this study has confirmed, that neuroblastomas are very heterogeneous; no single genetic change common to all neuroblastomas has yet been identified. One genetic aberration found frequently in this pediatric tumor is MYCN gene amplification. Recently we identified a new subset of tumors showing MYCN gain (small increases in gene number arising from unbalanced translocation). To investigate whether gain precedes amplification or is an independent event, we surveyed 200 primary tumors for MYCN copy number with fluorescence in situ hybridization; 152 of 200 (76%) were MYCN single-copy tumors, whereas 48 of 200 (24%) tumors harbored MYCN abnormalities: 36 of the 48 (75%) had MYCN amplification and 12 (25%) had MYCN gain. Among the 36 with MYCN amplified gene, we found four that also showed gain. In three tumors exhibiting simultaneous gain and amplification, these two events were detected in neighboring cells. In the fourth case we detected only MYCN gain in metastatic neuroblasts in the bone marrow, but both MYCN amplification and gain in the primary tumor. The detailed study of these four cases suggests that there may be several different mechanisms leading to increase in MYCN copy number. Further studies in other human malignancies are necessary to determine whether simultaneous gain and amplification are specific to neuroblastoma or constitute a general mechanism by which tumor cells can acquire selective growth advantage.


The Conundrum Posed by Cellular Heterogeneity in Analysis of Human Neuroblastoma

September 2004

·

12 Reads

·

13 Citations

JNCI Journal of the National Cancer Institute

Examining the methylation patterns of gene promoters has become a powerful tool in the effort to identify and distinguish different tumor subtypes (1,2). Typically associated with tumor suppressor genes, epigenetic silencing of gene expression has been seen in several different cancers. Aberrant hypermethyl- ation has been recorded for genes that function in various aspects of cancer biology, including cell cycle regulation, tumor sup- pression, apoptosis, and metastasis. The article by Alaminos et al. (3) in this issue is a clear extension of this previous research. DNA methylation silences gene expression through the ad- dition of methyl groups to cytosine residues of CpG-rich islands present in the promoter region of genes. Whereas housekeeping genes possess CpG regions that are typically unmethylated and therefore transcriptionally active in all cell types, tissue-specific genes are unmethylated in cells actively expressing the genes and are normally methylated in all other differentiated tissues. Patterns of silencing are specified during development; methyl- ation is essential for normal imprinting, X-chromosome inacti- vation, and differential gene expression during embryonic growth. Changes in DNA methylation patterns have recently been associated with specific cancers, and CpG island hyper- methylation profiles for primary tumors have become accepted as a common means to distinguish human cancer subtypes (1,2). Alaminos et al. (3) initially chose 45 candidate genes repre- sentative of important cellular processes and compared their methylation states in 10 human neuroblastoma cell lines and clones by using methylation-specific polymerase chain reaction. The cell lines comprise homogeneous populations of the various cell types—that is, S-type (substrate adherent), N-type (neuro- blastic), and I-type (stem) cells— derived from tumors and were used to determine whether specific differences in the methyl- ation profiles of the candidate genes were associated with cell phenotype. I-type cells can differentiate into either neuroblasts


Citations (78)


... The ELAV family of RNAbinding proteins has gained significant attention due to their potential as therapeutic targets for cancer treatment, as well as their ability to regulate a wide range of RNA targets post-transcriptionally [40][41][42] . Previous studies have reported that ELAVL4 binds to the 3'-UTR of MYCN mRNA in neuroblastoma [43][44][45] . However, our analysis found that ELAVL3 was highly expressed in NEPC through the combination of multiple RNA-seq datasets from prostate cancer patients (Fig. 1a). ...

Reference:

The ELAVL3/MYCN positive feedback loop provides a therapeutic target for neuroendocrine prostate cancer
Reciprocal antagonistic regulation of N-myc mRNA by miR‑17 and the neuronal-specific RNA-binding protein HuD
  • Citing Article
  • May 2017

Oncology Reports

... ϫ300. The karyotypes of these two cell lines were consistent with previous reports (22), and the similarities between them, i.e., del(1p), der(3)t(3p;17q), der (4)t(1q;4q), suggest that they originated from the same tumor. The double minute chromosomes in SKNBE(1n) and HSRs in SKNBE(2c) have previously been shown to be sites of MYCN amplification, and the progression from double minute chromosomes to HSRs previously was recorded (23). ...

Chromosomal and biochemical properties of human neuroblastoma lines and clones in cell culture
  • Citing Article
  • January 1979

... Because several other systems have demonstrated a relationship between unstable drug resistance and the presence within the nucleus of small (-0.5 ,um) (5), usually paired, chromatin-positive structures termed DMs (7,29,40), karyotype analysis was carried out on the transformants and amplified transformants in comparison with their parental drug-sensitive lines. DMs were observed in drug-resistant DNA transformants and in greater numbers in the amplified transformants (Fig. 3, Table 2). ...

Vincristine-resistant human neuroblastoma cells have double minutes (DMs) and homogeneously staining regions (HSRs)
  • Citing Article
  • January 1981

... This differential representation of Myc/Pvt1 amplified sequences in this tumor may be due to the disappearance of unstable dmin and subsequent integration into the specific chromosomal site to yield an hsr in the metastatic tumor. This interconvertible phenomenon has previously been reported in several cancer types (Levan and Levan, 1982;Biedler et al., 1983;Van Hoff et al., 1990). Recently, L'Abbate et al. ...

Homogeneously Staining Regions and Double Minute Chromosomes, Prevalent Cytogenetic Abnormalities of Human Neuroblastoma Cells
  • Citing Chapter
  • December 1983

... The current study characterizes antibodies raised against determinants on a multidrug-resistant subline (SH-SY5Y/ VCR) selected with vincristine from explanted human tumor cells of neural crest origin. SH-SY5Y/VCR neuroblastoma cells show a 1400-fold increase in resistance to the selective agent and cross-resistance to Adriamycin and actinomycin D (12) and are known to have amplified Pgp genes and increased Pgp expression (13). Three antibodies (HYB-241, HYB-612, and HYB-195) were selected for initial characterization on the basis of ELISA results showing increased reactivity to drug-resistant, as compared to sensitive, cells. ...

Cellular Concomitants of Multidrug Resistance
  • Citing Chapter
  • December 1988

... In the early 1980s, the first karyotypic reports of NB 3 cell lines and stage 4 tumors revealed several nonrandom chromosomal abnormalities associated with the disease including losses on chromosomes 1p and 11q, gains on chromosomes 17q and 1q, the presence of homozygous staining regions and double minutes, and changes in the normal diploid chromosomal content (1)(2)(3)(4)(5). Amplification of the proto-oncogene MYCN was subsequently identified in the homozygous staining regions and double minutes (6). ...

Human neuroblastoma cytogenetics: Search for significance of homogeneously staining regions and double minute chromosomes
  • Citing Article
  • January 1980

... Presently, two hypotheses, genetic and epigenetic, have been proposed to explain mechanisms of acquired cancer drug resistance (13 -16). ''Genetic'' is defined as a heritable change in the DNA sequence, and according to this mechanism, the occurrence of random drug-induced mutational events leads to the formation of drug-resistant cells from sensitive cells (13,14). ''Epigenetic'' refers to the information contained in chromatin rather than in the actual DNA sequence (17) and consists of DNA methylation and histone modifications. ...

Genetics of drug resistance
  • Citing Article
  • May 1995

Cancer Treatment and Research

... In the present study, N-and S-type cells were enriched from the parental SH-SY5Y neuroblastoma cell line which, although mainly composed of N-type cells, S-type cells remain present due to the ability of cells to transdifferentiate between cell phenotypes [7,35]. Cell populations were induced to differentiate by the addition of 9cRA and characterised morphologically and biochemically using the neuronal marker proteins β-tubulin III and Bcl-2 [36][37][38][39] and the nonneuronal marker protein vimentin [3]. ...

Morphological interconversion of human neuroblastoma cells
  • Citing Article
  • January 1975

In Vitro Cellular & Developmental Biology - Plant

... Her iki hücre tipini oluşturabilme ve kendini yenileme özelliğine sahip olduğundan kök hücre olarak karakterize edilmiştir. 12 Normal nöronal gelişim sırasında nöral progenitör hücrelerin polarite kaybı ve asimetrik bölünmesinin nöroblastom kök hücresini meydana getirdiği Drosophila melanogaster ile yapılan çalışmada gösterilmiştir. 13 SPDYA, asimetrik bölünmeyi kontrol ederek nöroblastom kök hücresi oluşumunu kontrol etmektedir. ...

Human neuroblastoma I-type cells are malignant neural crest stem cells.
  • Citing Article
  • April 1995

Cell growth & differentiation: the molecular biology journal of the American Association for Cancer Research