Try a new search

Format these results:

Searched for:

in-biosketch:yes

person:hernae07

Total Results:

149


Clinical relevance of SKP2 alterations in metastatic melanoma

Rose, Amy E; Wang, Guimin; Hanniford, Douglas; Monni, Stefano; Tu, Ting; Shapiro, Richard L; Berman, Russell S; Pavlick, Anna C; Pagano, Michele; Darvishian, Farbod; Mazumdar, Madhu; Hernando, Eva; Osman, Iman
In this study, we investigated the mechanism(s) of altered expression of protooncogene SKP2 in metastatic melanoma and its clinical relevance in patients with metastatic melanoma. The genomic status of SKP2 was assessed in cell lines by sequencing, single nucleotide polymorphism array, and genomic PCR. Copy number status was then evaluated for concordance with SKP2 mRNA and protein expression. SKP2 protein was further evaluated by immunohistochemistry in 93 human metastatic tissues. No mutations were identified in SKP2. Increased copy number at the SKP2 locus was observed in 6/14 (43%) metastatic cell lines and in 9/22 (41%) human metastatic tissues which was associated with overexpression of SKP2 protein. Overexpression of SKP2 protein in human tissues was associated with worse survival in a multivariate model controlling for the site of metastasis. Copy number gain is a major contributing mechanism of SKP2 overexpression in metastatic melanoma. Results may have implications for the development of therapeutics that target SKP2
PMCID:3341662
PMID: 20883453
ISSN: 1755-148x
CID: 138133

HMGA2 overexpression-induced ovarian surface epithelial transformation is mediated through regulation of EMT genes

Wu, Jingjing; Liu, Zhaojian; Shao, Changshun; Gong, Yaoqin; Hernando, Eva; Lee, Peng; Narita, Masashi; Muller, William; Liu, Jinsong; Wei, Jian-Jun
The AT-hook transcription factor HMGA2 is an oncogene involved in the tumorigenesis of many malignant neoplasms. HMGA2 overexpression is common in both early and late-stage high-grade ovarian serous papillary carcinoma. To test whether HMGA2 participates in the initiation of ovarian cancer and promotion of aggressive tumor growth, we examined the oncogenic properties of HMGA2 in ovarian surface epithelial (OSE) cell lines. We found that introduction of HMGA2 overexpression was sufficient to induce OSE transformation in vitro. HMGA2-mediated OSE transformation resulted in tumor formation in the xenografts of nude mice. By silencing HMGA2 in HMGA2-overexpressing OSE and ovarian cancer cell lines, the aggressiveness of tumor cell growth behaviors was partially suppressed. Global gene profiling analyses revealed that HMGA2-mediated tumorigenesis was associated with expression changes of target genes and microRNAs that are involved in epithelial-to-mesenchymal transition (EMT). Lumican, a tumor suppressor that inhibits EMT, was found to be transcriptionally repressed by HMGA2 and was frequently lost in human high-grade serous papillary carcinoma. Our findings show that HMGA2 overexpression confers a powerful oncogenic signal in ovarian cancers through the modulation of EMT genes
PMCID:4434602
PMID: 21224353
ISSN: 1538-7445
CID: 133176

Correction: The Novel Gamma Secretase Inhibitor RO4929097 Reduces the Tumor Initiating Potential of Melanoma [Correction]

Huynh, Chanh; Poliseno, Laura; Segura, Miguel F; Medicherla, Ratna; Haimovic, Adele; Menendez, Silvia; Shang, Shulian; Pavlick, Anna; Shao, Yongzhao; Darvishian, Farbod; Boylan, John F; Osman, Iman; Hernando, Eva
[This corrects the article on p. e25264 in vol. 6.]
PMCID:3207494
ORIGINAL:0007335
ISSN: 1932-6203
CID: 141637

The Novel Gamma Secretase Inhibitor RO4929097 Reduces the Tumor Initiating Potential of Melanoma

Huynh, Chanh; Poliseno, Laura; Segura, Miguel F; Medicherla, Ratna; Haimovic, Adele; Menendez, Silvia; Shang, Shulian; Pavlick, Anna; Shao, Yongzhao; Darvishian, Farbod; Boylan, John F; Osman, Iman; Hernando, Eva
Several reports have demonstrated a role for aberrant NOTCH signaling in melanoma genesis and progression, prompting us to explore if targeting this pathway is a valid therapeutic approach against melanoma. We targeted NOTCH signaling using RO4929097, a novel inhibitor of gamma secretase, which is a key component of the enzymatic complex that cleaves and activates NOTCH. The effects of RO4929097 on the oncogenic and stem cell properties of a panel of melanoma cell lines were tested both in vitro and in vivo, using xenograft models. In human primary melanoma cell lines, RO4929097 decreased the levels of NOTCH transcriptional target HES1. This was accompanied by reduced proliferation and impaired ability to form colonies in soft agar and to organize in tridimensional spheres. Moreover, RO4929097 affected the growth of human primary melanoma xenograft in NOD/SCID/IL2gammaR-/- mice and inhibited subsequent tumor formation in a serial xenotransplantation model, suggesting that inhibition of NOTCH signaling suppresses the tumor initiating potential of melanoma cells. In addition, RO4929097 decreased tumor volume and blocked the invasive growth pattern of metastatic melanoma cell lines in vivo. Finally, increased gene expression of NOTCH signaling components correlated with shorter post recurrence survival in metastatic melanoma cases. Our data support NOTCH inhibition as a promising therapeutic strategy against melanoma
PMCID:3182998
PMID: 21980408
ISSN: 1932-6203
CID: 138712

MicroRNA-130b contributes to mesenchymal differentiation and leiomyosarcomagenesis [Meeting Abstract]

Danielson, Laura; Guijarro, Maria; Zavadil, Jiri; Wei, Jianjun; Levine, Douglas; Hernando, Eva
ISI:000209701306048
ISSN: 1538-7445
CID: 2225422

The histone variant macroH2A suppresses melanoma progression through regulation of CDK8

Kapoor, Avnish; Goldberg, Matthew S; Cumberland, Lara K; Ratnakumar, Kajan; Segura, Miguel F; Emanuel, Patrick O; Menendez, Silvia; Vardabasso, Chiara; Leroy, Gary; Vidal, Claudia I; Polsky, David; Osman, Iman; Garcia, Benjamin A; Hernando, Eva; Bernstein, Emily
Cancer is a disease consisting of both genetic and epigenetic changes. Although increasing evidence demonstrates that tumour progression entails chromatin-mediated changes such as DNA methylation, the role of histone variants in cancer initiation and progression currently remains unclear. Histone variants replace conventional histones within the nucleosome and confer unique biological functions to chromatin. Here we report that the histone variant macroH2A (mH2A) suppresses tumour progression of malignant melanoma. Loss of mH2A isoforms, histone variants generally associated with condensed chromatin and fine-tuning of developmental gene expression programs, is positively correlated with increasing malignant phenotype of melanoma cells in culture and human tissue samples. Knockdown of mH2A isoforms in melanoma cells of low malignancy results in significantly increased proliferation and migration in vitro and growth and metastasis in vivo. Restored expression of mH2A isoforms rescues these malignant phenotypes in vitro and in vivo. We demonstrate that the tumour-promoting function of mH2A loss is mediated, at least in part, through direct transcriptional upregulation of CDK8. Suppression of CDK8, a colorectal cancer oncogene, inhibits proliferation of melanoma cells, and knockdown of CDK8 in cells depleted of mH2A suppresses the proliferative advantage induced by mH2A loss. Moreover, a significant inverse correlation between mH2A and CDK8 expression levels exists in melanoma patient samples. Taken together, our results demonstrate that mH2A is a critical component of chromatin that suppresses the development of malignant melanoma, a highly intractable cutaneous neoplasm
PMCID:3057940
PMID: 21179167
ISSN: 1476-4687
CID: 134348

New gamma-secretase inhibitor (RO4929097) targeting notch signaling in melanoma [Meeting Abstract]

Osman, I; Poliseno, L; Huynh, C T; Segura, M F; Medicherla, R; Menendez, S; Rose, A E; Pavlick, A C; Boylan, J; Hernando, E
The incidence of melanoma has increased 3-7% per year, and is now approaching 30 per 100,000 individuals. This rate is faster than any other cancer, and is predicted to double every 10-20 years [1]. Surgery can be curative in Stage I, II, or III disease, but 75% of patients with deep primary lesions will develop extensive recurrence or distant metastases (Stage IV disease). To date, no curative treatment exists for Stage IV melanoma and these patients have a median overall survival of only 7 months [2]. A subset of patients can be salvaged with surgical resection of metastatic sites, but no adjuvant therapy has further improved the outcome [3]. The only FDA approved adjuvant therapy for Stage IV melanoma is alpha-interferon. Several trials have demonstrated an increase in relapse-free survival; however, toxicity is high and overall survival remains controversial. Several reports have demonstrated a role for aberrant Notch signaling in melanoma genesis or progression, prompting us to explore if targeting this pathway is a valid therapeutic approach against melanoma. To investigate the potential benefits of Notch inhibition in melanoma, we are using RO4929097, a novel inhibitor of gamma secretase, a key component of the enzymatic complex that cleaves and activates Notch. RO4929097 is completing Phase I dose escalation in cancer patients and a CTEP-sponsored Phase II clinical trial in melanoma is currently under review. We have generated DNA microarray data for a series of 22 melanoma cell lines at both the gene expression and DNA copy number level. Preliminary gene expression analysis has indicated that melanoma cells over express crucial components of NOTCH-pathway, such as Hey1 Hey2 and NOXA. Furthermore, integration of gene expression and copy number data for NOTCH2 suggests that the increased DNA copy number play a role in the increase in gene expression. We have also tested the efficacy of RO4929047 in human melanoma cell lines. We have observed that treatment with RO4929097 for 24h causes a!
EMBASE:71298305
ISSN: 1527-2729
CID: 783372

MIR-7 AND MIR-130B ARE DIFFERENTIALLY REGULATED DURING MESENCHYMAL STEM CELL COMMITMENT [Meeting Abstract]

Palmer, G.; Danielson, L.; Attur, M.; Abramson, S. B.; Hernando, E.
ISI:000283452900096
ISSN: 1063-4584
CID: 120554

Profiling and functional analyses of microRNAs and their target gene products in human uterine leiomyomas

Zavadil, Jiri; Ye, Huihui; Liu, Zhaojian; Wu, JingJing; Lee, Peng; Hernando, Eva; Soteropoulos, Patricia; Toruner, Gokce A; Wei, Jian-Jun
BACKGROUND: Human uterine leiomyomas (ULM) are characterized by dysregulation of a large number of genes and non-coding regulatory microRNAs. In order to identify microRNA::mRNA associations relevant to ULM pathogenesis, we examined global correlation patterns between the altered microRNA expression and the predicted target genes in ULMs and matched myometria. METHODOLOGY/PRINCIPAL FINDINGS: Patterns of inverse association of microRNA with mRNA expression in ULMs revealed an involvement of multiple candidate pathways, including extensive transcriptional reprogramming, cell proliferation control, MAP kinase, TGF-beta, WNT, JAK/STAT signaling, remodeling of cell adhesion, and cell-cell and cell-matrix contacts. We further examined the correlation between the expression of the selected target gene protein products and microRNAs in thirty-six paired sets of leiomyomas and matched myometria. We found that a number of dysregulated microRNAs were inversely correlated with their targets at the protein level. The comparative genomic hybridization (CGH) in eight ULM patients revealed that partially shared deletions of two distinct chromosomal regions might be responsible for loss of cancer-associated microRNA expression and could thus contribute to the ULM pathogenesis via deregulation of target mRNAs. Last, we functionally tested the repressor effects of selected cancer-related microRNAs on their predicted target genes in vitro. CONCLUSIONS/SIGNIFICANCE: We found that some but not all of the predicted and inversely correlated target genes in ULMs can be directly regulated by microRNAs in vitro. Our findings provide a broad overview of molecular events underlying the tumorigenesis of uterine ULMs and identify select genetic and regulatory events that alter microRNA expression and may play important roles in ULM pathobiology by positively regulating tumor growth while maintaining the non-invasive character of ULMs
PMCID:2927438
PMID: 20808773
ISSN: 1932-6203
CID: 112544

A Differentiation-Based MicroRNA Signature Identifies Leiomyosarcoma as a Mesenchymal Stem Cell-Related Malignancy

Danielson, Laura S; Menendez, Silvia; Attolini, Camille Stephan-Otto; Guijarro, Maria V; Bisogna, Maria; Wei, Jianjun; Socci, Nicholas D; Levine, Douglas A; Michor, Franziska; Hernando, Eva
Smooth muscle (SM) is a spontaneously contractile tissue that provides physical support and function to organs such as the uterus. Uterine smooth muscle-related neoplasia comprise common well-differentiated benign lesions called leiomyomas (ULM), and rare, highly aggressive and pleomorphic tumors named leiomyosarcomas (ULMS). MicroRNAs (miRNAs) are small non-coding RNAs that play essential roles in normal cellular development and tissue homeostasis that can be used to accurately subclassify different tumor types. Here, we demonstrate that miRNAs are required for full smooth muscle cell (SMC) differentiation of bone marrow-derived human mesenchymal stem cells (hMSCs). We also report a miRNA signature associated with this process. Moreover, we show that this signature, along with miRNA profiles for ULMS and ULM, are able to subclassify tumors of smooth muscle origin along SM differentiation. Using multiple computational analyses, we determined that ULMS are more similar to hMSCs as opposed to ULM, which are linked with more mature SMCs and myometrium. Furthermore, a comparison of the SM differentiation and ULMS miRNA signatures identified miRNAs strictly associated with SM maturation or transformation, as well as those modulated in both processes indicating a possible dual role. These results support separate origins and/or divergent transformation pathways for ULM and ULMS, resulting in drastically different states of differentiation. In summary, this work expands on our knowledge of the regulation of SM differentiation and sarcoma pathogenesis
PMCID:2913343
PMID: 20558575
ISSN: 1525-2191
CID: 111539