Try a new search

Format these results:

Searched for:

person:hernae07

in-biosketch:yes

Total Results:

143


circSamd4 represses myogenic transcriptional activity of PUR proteins

Pandey, Poonam R; Yang, Jen-Hao; Tsitsipatis, Dimitrios; Panda, Amaresh C; Noh, Ji Heon; Kim, Kyoung Mi; Munk, Rachel; Nicholson, Thomas; Hanniford, Douglas; Argibay, Diana; Yang, Xiaoling; Martindale, Jennifer L; Chang, Ming-Wen; Jones, Simon W; Hernando, Eva; Sen, Payel; De, Supriyo; Abdelmohsen, Kotb; Gorospe, Myriam
By interacting with proteins and nucleic acids, the vast family of mammalian circRNAs is proposed to influence many biological processes. Here, RNA sequencing analysis of circRNAs differentially expressed during myogenesis revealed that circSamd4 expression increased robustly in mouse C2C12 myoblasts differentiating into myotubes. Moreover, silencing circSamd4, which is conserved between human and mouse, delayed myogenesis and lowered the expression of myogenic markers in cultured myoblasts from both species. Affinity pulldown followed by mass spectrometry revealed that circSamd4 associated with PURA and PURB, two repressors of myogenesis that inhibit transcription of the myosin heavy chain (MHC) protein family. Supporting the hypothesis that circSamd4 might complex with PUR proteins and thereby prevent their interaction with DNA, silencing circSamd4 enhanced the association of PUR proteins with the Mhc promoter, while overexpressing circSamd4 interfered with the binding of PUR proteins to the Mhc promoter. These effects were abrogated when using a mutant circSamd4 lacking the PUR binding site. Our results indicate that the association of PUR proteins with circSamd4 enhances myogenesis by contributing to the derepression of MHC transcription.
PMID: 31980816
ISSN: 1362-4962
CID: 4274142

Epigenetic Silencing of CDR1as Drives IGF2BP3-Mediated Melanoma Invasion and Metastasis

Hanniford, Douglas; Ulloa-Morales, Alejandro; Karz, Alcida; Berzoti-Coelho, Maria Gabriela; Moubarak, Rana S; Sánchez-Sendra, Beatriz; Kloetgen, Andreas; Davalos, Veronica; Imig, Jochen; Wu, Pamela; Vasudevaraja, Varshini; Argibay, Diana; Lilja, Karin; Tabaglio, Tommaso; Monteagudo, Carlos; Guccione, Ernesto; Tsirigos, Aristotelis; Osman, Iman; Aifantis, Iannis; Hernando, Eva
Metastasis is the primary cause of death of cancer patients. Dissecting mechanisms governing metastatic spread may uncover important tumor biology and/or yield promising therapeutic insights. Here, we investigated the role of circular RNAs (circRNA) in metastasis, using melanoma as a model aggressive tumor. We identified silencing of cerebellar degeneration-related 1 antisense (CDR1as), a regulator of miR-7, as a hallmark of melanoma progression. CDR1as depletion results from epigenetic silencing of LINC00632, its originating long non-coding RNA (lncRNA) and promotes invasion in vitro and metastasis in vivo through a miR-7-independent, IGF2BP3-mediated mechanism. Moreover, CDR1as levels reflect cellular states associated with distinct therapeutic responses. Our study reveals functional, prognostic, and predictive roles for CDR1as and expose circRNAs as key players in metastasis.
PMID: 31935372
ISSN: 1878-3686
CID: 4263262

Melanoma-secreted amyloid beta supresses neuroinflammation and promotes brain metastasis [Meeting Abstract]

Kleffman, K; Levinson, G; Dhabaria, A; Galan, F; Wong, E; Itter, R V; De, Miera E; Tranos, J; Osman, I; Li, Y; Ueberheide, B; Liddelow, S; Ruggles, K; Schneider, R; Hernando, E
Melanoma brain metastasis is the largest cause of melanoma morbidity and mortality, and melanoma has the highest rate of brain metastasis of any cancer. The mechanisms that mediate melanoma brain metastasis remain poorly understood. We characterized patient-derived Short-Term Cultures (STCs) as a novel model system for the study of melanoma brain metastasis. Unbiased proteomics analysis of STCs revealed striking alterations in brain metastasis vs non-brain metastasis derived STCs in proteins related to neurodegeneration. Through in-vivo assays, we show that loss of Amyloid Precursor Protein (APP) in melanoma cells dramatically inhibits melanoma brain metastasis formation without affecting metastasis to other organs and that amyloid beta is the form of APP critically required for melanoma brain metastasis. Additionally, we demonstrate that APP is required for late growth and survival of melanoma cells in the brain parenchyma. Furthermore, we demonstrate that melanoma-derived amyloid beta polarizes astrocytes to an anti-inflammatory secretory phenotype that inhibits microglial phagocytosis of melanoma cells. Finally, we show that treatment of mice with a beta secretase inhibitor (LY2886721), which prevents amyloid beta production, decreases brain metastatic burden. Our results demonstrate a critical role for amyloid beta in melanoma brain metastasis, establish a novel connection between brain metastasis and neurodegenerative pathologies, and show that amyloid beta is a promising therapeutic target for brain metastasis treatment. Studies to further characterize how amyloid beta acts in the melanoma brain metastasis microenvironment are currently underway
EMBASE:631885213
ISSN: 1755-148x
CID: 4471292

Primary Melanoma Histologic Subtype: Impact on Survival and Response to Therapy

Lattanzi, Michael; Lee, Yesung; Simpson, Danny; Moran, Una; Darvishian, Farbod; Kim, Randie H; Hernando, Eva; Polsky, David; Hanniford, Doug; Shapiro, Richard; Berman, Russell; Pavlick, Anna C; Wilson, Melissa A; Kirchhoff, Tomas; Weber, Jeffrey S; Zhong, Judy; Osman, Iman
Background/UNASSIGNED:Two primary histologic subtypes, superficial spreading melanoma (SSM) and nodular melanoma (NM), comprise the majority of all cutaneous melanomas. NM is associated with worse outcomes, which have been attributed to increased thickness at presentation, and it is widely expected that NM and SSM would exhibit similar behavior once metastasized. Herein, we tested the hypothesis that primary histologic subtype is an independent predictor of survival and may impact response to treatment in the metastatic setting. Methods/UNASSIGNED:We examined the most recent Surveillance, Epidemiology, and End Results (SEER) cohort (n = 118 508) and the New York University (NYU) cohort (n = 1621) with available protocol-driven follow-up. Outcomes specified by primary histology were studied in both the primary and metastatic settings with respect to BRAF-targeted therapy and immunotherapy. We characterized known driver mutations and examined a 140-gene panel in a subset of NM and SSM cases using next-generation sequencing. All statistical tests were two-sided. Results/UNASSIGNED:NM was an independent risk factor for death in both the SEER (hazard ratio [HR] = 1.55, 95% confidence interval [CI] = 1.41 to 1.70, P < .001) and NYU (HR = 1.47, 95% CI = 1.05, 2.07, P = .03) cohorts, controlling for thickness, ulceration, stage, and other variables. In the metastatic setting, NM remained an independent risk factor for death upon treatment with BRAF-targeted therapy (HR = 3.33, 95% CI = 1.06 to 10.47, P = .04) but showed no statistically significant difference with immune checkpoint inhibition. NM was associated with a higher rate of NRAS mutation (P < .001), and high-throughput sequencing revealed NM-specific genomic alterations in NOTCH4, ANK3, and ZNF560, which were independently validated. Conclusions/UNASSIGNED:Our data reveal distinct clinical and biological differences between NM and SSM that support revisiting the prognostic and predictive impact of primary histology subtype in the management of cutaneous melanoma.
PMID: 29912415
ISSN: 1460-2105
CID: 3158042

A Leukocyte Infiltration Score Defined by a Gene Signature Predicts Melanoma Patient Prognosis

Zhao, Yanding; Schaafsma, Evelien; Gorlov, Ivan P; Hernando, Eva; Thomas, Nancy E; Shen, Ronglai; Turk, Mary Jo; Berwick, Marianne; Amos, Christopher I; Cheng, Chao
: Melanoma is the most aggressive type of skin cancer in the United States with an increasing incidence. Melanoma lesions often exhibit high immunogenicity, with infiltrating immune cells playing important roles in regression of tumors occurring spontaneously or caused by therapeutic treatment. Computational and experimental methods have been used to estimate the abundance of immune cells in tumors, but their applications are limited by the requirement of large gene sets or multiple antibodies. Although the prognostic role of immune cells has been appreciated, a systematic investigation of their association with clinical factors, genomic features, prognosis and treatment response in melanoma is still lacking. This study, identifies a 25-gene signature based on RNA-seq data from The Cancer Genome Atlas (TCGA)-Skin Cutaneous Melanoma (TCGA-SKCM) dataset. This signature was used to calculate sample-specific Leukocyte Infiltration Scores (LIS) in six independent melanoma microarray datasets and scores were found to vary substantially between different melanoma lesion sites and molecular subtypes. For metastatic melanoma, LIS was prognostic in all datasets with high LIS being associated with good survival. The current approach provided additional prognostic information over established clinical factors, including age, tumor stage, and gender. In addition, LIS was predictive of patient survival in stage III melanoma, and treatment efficacy of tumor-specific antigen vaccine. IMPLICATIONS: This study identifies a 25-gene signature that effectively estimates the level of immune cell infiltration in melanoma, which provides a robust biomarker for predicting patient prognosis.
PMID: 30171176
ISSN: 1557-3125
CID: 3562642

Characterization of MicroRNAs Regulating FOXO Expression

Hanniford, Doug; Hernando, Eva
MicroRNAs are critical post-transcriptional regulators of a majority of genes, of which the FOXO family of transcription factors is no exception. Here, we describe generalizable methods, including 3' UTR reporter assays and western blotting after microRNA manipulation, to test if a candidate miRNA (miR-182) directly targets a candidate (FOXO3) gene product. We also provide guidance on candidate miRNA selection and unbiased miRNA-target identification methods.
PMID: 30414141
ISSN: 1940-6029
CID: 3458362

Identification of gene expression levels in primary melanoma associated with clinically meaningful characteristics

Gorlov, Ivan; Orlow, Irene; Ringelberg, Carol; Hernando, Eva; Ernstoff, Marc S; Cheng, Chao; Her, Stephanie; Parker, Joel S; Thompson, Cheryl L; Gerstenblith, Meg R; Berwick, Marianne; Amos, Christopher
Factors influencing melanoma survival include sex, age, clinical stage, lymph node involvement, as well as Breslow thickness, presence of tumor-infiltrating lymphocytes based on histological analysis of primary melanoma, mitotic rate, and ulceration. Identification of genes whose expression in primary tumors is associated with these key tumor/patient characteristics can shed light on molecular mechanisms of melanoma survival. Here, we show results from a gene expression analysis of formalin-fixed paraffin-embedded primary melanomas with extensive clinical annotation. The Cancer Genome Atlas data on primary melanomas were used for validation of nominally significant associations. We identified five genes that were significantly associated with the presence of tumor-infiltrating lymphocytes in the joint analysis after adjustment for multiple testing: IL1R2, PPL, PLA2G3, RASAL1, and SGK2. We also identified two genes significantly associated with melanoma metastasis to the regional lymph nodes (PIK3CG and IL2RA), and two genes significantly associated with sex (KDM5C and KDM6A). We found that LEF1 was significantly associated with Breslow thickness and CCNA2 and UBE2T with mitosis. RAD50 was the gene most significantly associated with survival, with a higher level of expression associated with worse survival.
PMID: 29975213
ISSN: 1473-5636
CID: 3186162

Lysyl oxidase-like 3 is required for melanoma cell survival by maintaining genomic stability

Santamaría, Patricia G; Floristán, Alfredo; Fontanals-Cirera, Bárbara; Vázquez-Naharro, Alberto; Santos, Vanesa; Morales, Saleta; Yuste, Lourdes; Peinado, Héctor; García-Gómez, Antonio; Portillo, Francisco; Hernando, Eva; Cano, Amparo
Lysyl oxidase-like 3 (LOXL3) is a member of the lysyl oxidase family comprising multifunctional enzymes with depicted roles in extracellular matrix maturation, tumorigenesis, and metastasis. In silico expression analyses followed by experimental validation in a comprehensive cohort of human cell lines revealed a significant upregulation of LOXL3 in human melanoma. We show that LOXL3 silencing impairs cell proliferation and triggers apoptosis in various melanoma cell lines. Further supporting a pro-oncogenic role in melanoma, LOXL3 favors tumor growth in vivo and cooperates with oncogenic BRAF in melanocyte transformation. Upon LOXL3 depletion, melanoma cells display a faulty DNA damage response (DDR), characterized by ATM checkpoint activation and inefficient ATR activation leading to the accumulation of double-strand breaks (DSBs) and aberrant mitosis. Consistent with these findings, LOXL3 binds to proteins involved in the maintenance of genome integrity, in particular BRCA2 and MSH2, whose levels dramatically decrease upon LOXL3 depletion. Moreover, LOXL3 is required for efficient DSB repair in melanoma cells. Our results reveal an unexpected role for LOXL3 in the control of genome stability and melanoma progression, exposing its potential as a novel therapeutic target in malignant melanoma, a very aggressive condition yet in need for more effective treatment options.
PMCID:5907912
PMID: 29229995
ISSN: 1476-5403
CID: 2844502

Differential phosphorylation and n-terminal configuration of capsid subunits in parvovirus assembly and viral trafficking

Gil-Ranedo, Jon; Hernando, Eva; Valle, Noelia; Riolobos, Laura; Maroto, Beatriz; Almendral, José M
The T1 parvovirus Minute Virus of Mice (MVM) was used to study the roles that phosphorylation and N-terminal domains (Nt) configuration of capsid subunits may play in icosahedral nuclear viruses assembly. In synchronous MVM infection, capsid subunits newly assembled as two types of cytoplasmic trimeric intermediates (3VP2, and 1VP1:2VP2) harbored a VP1 phosphorylation level fivefold higher than that of VP2, and hidden Nt. Upon nuclear translocation at S phase, VP1-Nt became exposed in the heterotrimer and subsequent subviral assembly intermediates. Empty capsid subunits showed a phosphorylation level restored to VP1:VP2 stoichiometry, and the Nt concealed in their interior. However ssDNA-filled virus maturing at S/G2 lacked VP1 phosphorylation and one major VP2 phosphopeptide, and exposed VP2-Nt. Endosomal VP2-Nt cleavage resulted in VP3 subunits devoid of any phospholabel, implying that incoming viral particles specifically harbor a low phosphorylation status. Phosphorylation provides a mechanistic coupling of parvovirus nuclear assembly to the cell cycle.
PMID: 29524834
ISSN: 1096-0341
CID: 3114362

miR-204-5p and miR-211-5p contribute to BRAF inhibitor resistance in melanoma

Díaz-Martínez, Marta; Benito-Jardón, Lucía; Alonso, Lola; Koetz-Ploch, Lisa; Hernando, Eva; Teixido, Joaquin
Melanoma treatment with the BRAF V600E inhibitor vemurafenib (VMF) provides therapeutic benefits but the common emergence of drug resistance remains a challenge. We generated A375 melanoma cells resistant to VMF with the goal of investigating changes in miRNA expression patterns that might contribute to resistance. Increased expression of miR-204-5p and miR-211-5p occurring in VMF-resistant cells was determined to impact VMF response. Their expression was rapidly affected by VMF treatment through RNA stabilization. Similar effects were elicited by MEK and ERK inhibitors but not AKT or Rac inhibitors. Ectopic expression of both miRNA in drug-naive human melanoma cells was sufficient to confer VMF resistance and more robust tumor growth in vivo. Conversely, silencing their expression in resistant cells inhibited cell growth. Joint overexpression of miR-204-5p and miR-211-5p durably stimulated Ras and MAPK upregulation after VMF exposure. Overall, our findings show how upregulation of miR-204-5p and miR-211-5p following VMF treatment enables the emergence of resistance, with potential implications for mechanism-based strategies to improve VMF responses.
PMCID:5815895
PMID: 29229605
ISSN: 1538-7445
CID: 2844552