Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Julia C. Cronin is active.

Publication


Featured researches published by Julia C. Cronin.


Nature Genetics | 2009

Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4

Todd D. Prickett; Neena S Agrawal; Xiaomu Wei; Kristin E Yates; Jimmy Lin; John R. Wunderlich; Julia C. Cronin; Pedro Cruz; Nisc Comparative Sequencing Program; Steven A. Rosenberg; Yardena Samuels

Tyrosine phosphorylation is important in signaling pathways underlying tumorigenesis. We performed a mutational analysis of the protein tyrosine kinase (PTK) gene family in cutaneous metastatic melanoma. We identified 30 somatic mutations affecting the kinase domains of 19 PTKs and subsequently evaluated the entire coding regions of the genes encoding these 19 PTKs for somatic mutations in 79 melanoma samples. We found ERBB4 mutations in 19% of individuals with melanoma and found mutations in two other kinases (FLT1 and PTK2B) in 10% of individuals with melanomas. We examined seven missense mutations in the most commonly altered PTK gene, ERBB4, and found that they resulted in increased kinase activity and transformation ability. Melanoma cells expressing mutant ERBB4 had reduced cell growth after shRNA-mediated knockdown of ERBB4 or treatment with the ERBB inhibitor lapatinib. These studies could lead to personalized therapeutics specifically targeting the kinases that are mutationally altered in individual melanomas.


Nature Genetics | 2009

Analysis of the matrix metalloproteinase family reveals that MMP8 is often mutated in melanoma

Lavanya H. Palavalli; Todd D. Prickett; John R. Wunderlich; Xiaomu Wei; Allison S. Burrell; Patricia Porter-Gill; Sean Davis; Chenwei Wang; Julia C. Cronin; Neena S Agrawal; Jimmy Lin; Wendy Westbroek; Shelley Hoogstraten-Miller; Alfredo A. Molinolo; Patricia Fetsch; Armando C. Filie; Michael P. O'Connell; Carolyn E. Banister; Jason Howard; Phillip Buckhaults; Ashani T. Weeraratna; Lawrence C. Brody; Steven A. Rosenberg; Yardena Samuels

A mutational analysis of the matrix metalloproteinase (MMP) gene family in human melanoma identified somatic mutations in 23% of melanomas. Five mutations in one of the most commonly mutated genes, MMP8, reduced MMP enzyme activity. Expression of wild-type but not mutant MMP8 in human melanoma cells inhibited growth on soft agar in vitro and tumor formation in vivo, suggesting that wild-type MMP-8 has the ability to inhibit melanoma progression.


Pigment Cell & Melanoma Research | 2009

Frequent Mutations in the MITF Pathway in Melanoma

Julia C. Cronin; John R. Wunderlich; Stacie K. Loftus; Todd D. Prickett; Xiaomu Wei; Katie Ridd; Swapna Vemula; Allison S. Burrell; Neena S Agrawal; Jimmy Lin; Carolyn E. Banister; Phillip Buckhaults; Steven A. Rosenberg; Boris C. Bastian; William J. Pavan; Yardena Samuels

Microphthalmia‐associated transcription factor (MITF) is involved in melanocyte cell development, pigmentation and neoplasia. To determine whether MITF is somatically mutated in melanoma, we compared the sequence of MITF from primary and metastatic lesions to patient‐matched normal DNA. In the 50 metastatic melanoma tumor lines analysed, we discovered four samples that had genomic amplifications of MITF and four that had MITF mutations in the regions encoding the transactivation, DNA binding or basic, helix‐loop‐helix domains. Sequence analysis for SOX10, a transcription factor, which both acts upstream of MITF and synergizes with MITF, identified an additional three samples with frameshift or nonsense mutations. Microphthalmia‐associated transcription factor and SOX10 were found to be mutated in a mutually exclusive fashion, possibly suggesting disruption in a common genetic pathway. Taken together we found that over 20% of the metastatic melanoma cases had alterations in the MITF pathway. We show that the MITF pathway is also altered in primary melanomas: 2/26 demonstrated mutations in MITF and 6/55 demonstrated mutations in SOX10. Our findings suggest that altered MITF function during melanomagenesis can be achieved by MITF amplification, MITF single base substitutions or by mutation of its regulator SOX10.


Cancer Research | 2008

Mutational Inactivation of PTPRD in Glioblastoma Multiforme and Malignant Melanoma

David A. Solomon; Jung-Sik Kim; Julia C. Cronin; Zita A. Sibenaller; Timothy C. Ryken; Steven A. Rosenberg; Habtom W. Ressom; Walter Jean; Darell D. Bigner; Hai Yan; Yardena Samuels; Todd Waldman

An additional tumor suppressor gene on chromosome 9p telomeric to the CDKN2A/B locus has long been postulated to exist. Using Affymetrix 250K single nucleotide polymorphism arrays to screen for copy number changes in glioblastoma multiforme (GBM), we detected a high frequency of deletions of the PTPRD gene, which encodes a receptor protein tyrosine phosphatase at chromosome 9p23-24.1. Missense and nonsense mutations of PTPRD were identified in a subset of the samples lacking deletions, including an inherited mutation with somatic loss of the wild-type allele. We then sequenced the gene in melanoma and identified 10 somatic mutations in 7 of 57 tumors (12%). Reconstitution of PTPRD expression in GBM and melanoma cells harboring deletions or mutations led to growth suppression and apoptosis that was alleviated by both the somatic and constitutional mutations. These data implicate PTPRD in the pathogenesis of tumors of neuroectodermal origin and, when taken together with other recent reports of PTPRD mutations in adenocarcinoma of the colon and lung, suggest that PTPRD may be one of a select group of tumor suppressor genes that are inactivated in a wide range of common human tumor types.


BMC Developmental Biology | 2011

SOX10 directly modulates ERBB3 transcription via an intronic neural crest enhancer

Megana K. Prasad; Xylena Reed; David U. Gorkin; Julia C. Cronin; Anthony R. McAdow; Kristopher Chain; Chani J. Hodonsky; Erin A. Jones; John Svaren; Anthony Antonellis; Stephen L. Johnson; Stacie K. Loftus; William J. Pavan; Andrew S. McCallion

BackgroundThe ERBB3 gene is essential for the proper development of the neural crest (NC) and its derivative populations such as Schwann cells. As with all cell fate decisions, transcriptional regulatory control plays a significant role in the progressive restriction and specification of NC derived lineages during development. However, little is known about the sequences mediating transcriptional regulation of ERBB3 or the factors that bind them.ResultsIn this study we identified three transcriptional enhancers at the ERBB3 locus and evaluated their regulatory potential in vitro in NC-derived cell types and in vivo in transgenic zebrafish. One enhancer, termed ERBB3_MCS6, which lies within the first intron of ERBB3, directs the highest reporter expression in vitro and also demonstrates epigenetic marks consistent with enhancer activity. We identify a consensus SOX10 binding site within ERBB3_MCS6 and demonstrate, in vitro, its necessity and sufficiency for the activity of this enhancer. Additionally, we demonstrate that transcription from the endogenous Erbb3 locus is dependent on Sox10. Further we demonstrate in vitro that Sox10 physically interacts with that ERBB3_MCS6. Consistent with its in vitro activity, we also show that ERBB3_MCS6 drives reporter expression in NC cells and a subset of its derivative lineages in vivo in zebrafish in a manner consistent with erbb3b expression. We also demonstrate, using morpholino analysis, that Sox10 is necessary for ERBB3_MCS6 expression in vivo in zebrafish.ConclusionsTaken collectively, our data suggest that ERBB3 may be directly regulated by SOX10, and that this control may in part be facilitated by ERBB3_MCS6.


Pigment Cell & Melanoma Research | 2014

Distinct microRNA expression signatures are associated with melanoma subtypes and are regulated by HIF1A

Hun-Way Hwang; Laura L. Baxter; Stacie K. Loftus; Julia C. Cronin; Niraj S. Trivedi; Bhavesh Borate; William J. Pavan

The complex genetic changes underlying metastatic melanoma need to be deciphered to develop new and effective therapeutics. Previously, genome‐wide microarray analyses of human melanoma identified two reciprocal gene expression programs, including transcripts regulated by either transforming growth factor, beta 1 (TGFβ1) pathways, or microphthalmia‐associated transcription factor (MITF)/SRY‐box containing gene 10 (SOX10) pathways. We extended this knowledge by discovering that melanoma cell lines with these two expression programs exhibit distinctive microRNA (miRNA) expression patterns. We also demonstrated that hypoxia‐inducible factor 1 alpha (HIF1A) is increased in TGFβ1 pathway‐expressing melanoma cells and that HIF1A upregulates miR‐210, miR‐218, miR‐224, and miR‐452. Reduced expression of these four miRNAs in TGFβ1 pathway‐expressing melanoma cells arrests the cell cycle, while their overexpression in mouse melanoma cells increases the expression of the hypoxic response gene Bnip3. Taken together, these data suggest that HIF1A may regulate some of the gene expression and biological behavior of TGFβ1 pathway‐expressing melanoma cells, in part via alterations in these four miRNAs.


Cancer Research | 2013

SOX10 Ablation Arrests Cell Cycle, Induces Senescence, and Suppresses Melanomagenesis

Julia C. Cronin; Dawn E. Watkins-Chow; Art Incao; Joanne H. Hasskamp; Nicola Schönewolf; Lauren G. Aoude; Nicholas K. Hayward; Boris C. Bastian; Reinhard Dummer; Stacie K. Loftus; William J. Pavan

The transcription factor SOX10 is essential for survival and proper differentiation of neural crest cell lineages, where it plays an important role in the generation and maintenance of melanocytes. SOX10 is also highly expressed in melanoma tumors, but a role in disease progression has not been established. Here, we report that melanoma tumor cell lines require wild-type SOX10 expression for proliferation and SOX10 haploinsufficiency reduces melanoma initiation in the metabotropic glutamate receptor 1 (Grm1(Tg)) transgenic mouse model. Stable SOX10 knockdown in human melanoma cells arrested cell growth, altered cellular morphology, and induced senescence. Melanoma cells with stable loss of SOX10 were arrested in the G1 phase of the cell cycle, with reduced expression of the melanocyte determining factor microphthalmia-associated transcription factor, elevated expression of p21WAF1 and p27KIP2, hypophosphorylated RB, and reduced levels of its binding partner E2F1. As cell-cycle dysregulation is a core event in neoplastic transformation, the role for SOX10 in maintaining cell-cycle control in melanocytes suggests a rational new direction for targeted treatment or prevention of melanoma.


PLOS ONE | 2018

Identification and functional analysis of SOX10 phosphorylation sites in melanoma

Julia C. Cronin; Stacie K. Loftus; Laura L. Baxter; Steve Swatkoski; Marjan Gucek; William J. Pavan

The transcription factor SOX10 plays an important role in vertebrate neural crest development, including the establishment and maintenance of the melanocyte lineage. SOX10 is also highly expressed in melanoma tumors, and SOX10 expression increases with tumor progression. The suppression of SOX10 in melanoma cells activates TGF-β signaling and can promote resistance to BRAF and MEK inhibitors. Since resistance to BRAF/MEK inhibitors is seen in the majority of melanoma patients, there is an immediate need to assess the underlying biology that mediates resistance and to identify new targets for combinatorial therapeutic approaches. Previously, we demonstrated that SOX10 protein is required for tumor initiation, maintenance and survival. Here, we present data that support phosphorylation as a mechanism employed by melanoma cells to tightly regulate SOX10 expression. Mass spectrometry identified eight phosphorylation sites contained within SOX10, three of which (S24, S45 and T240) were selected for further analysis based on their location within predicted MAPK/CDK binding motifs. SOX10 mutations were generated at these phosphorylation sites to assess their impact on SOX10 protein function in melanoma cells, including transcriptional activation on target promoters, subcellular localization, and stability. These data further our understanding of SOX10 protein regulation and provide critical information for identification of molecular pathways that modulate SOX10 protein levels in melanoma, with the ultimate goal of discovering novel targets for more effective combinatorial therapeutic approaches for melanoma patients.


Journal of Investigative Dermatology | 2018

Mutated MITF-E87R in Melanoma Enhances Tumor Progression via S100A4

Alice Nordlinger; Shani Dror; Abdel G. Elkahloun; Justine Del Rio; Elisa Stubbs; Tami Golan; Hagar Malcov; Todd D. Pricket; Julia C. Cronin; Shivang Parikh; Sapir Labes; Laetitia Thomas; Gal Yankovitz; Yuval Tabach; Carmit Levy; Yardena Samuels; Mehdi Khaled

Melanoma, a melanocyte origin neoplasm, is the most lethal type of skin cancer, and incidence is increasing. Several familial and somatic mutations have been identified in the gene encoding the melanocyte lineage master regulator, MITF; however, the neoplastic mechanisms of these mutant MITF variants are mostly unknown. Here, by performing unbiased analysis of the transcriptomes in cells expressing mutant MITF, we identified calcium-binding protein S100A4 as a downstream target of MITF-E87R. By using wild-type and mutant MITF melanoma lines, we found that both endogenous wild-type and MITF-E87R variants occupy the S100A4 promoter. Remarkably, whereas wild-type MITF represses S100A4 expression, MITF-E87R activates its transcription. The opposite effects of wild-type and mutant MITF result in opposing cellular phenotypes, because MITF-E87R via S100A4 enhanced invasion and reduced adhesion in contrast to wild-type MITF activity. Finally, we found that melanoma patients with altered S100A4 expression have poor prognosis. These data show that a change in MITF transcriptional activity from repression to activation of S100A4 that results from a point mutation in MITF alters melanoma invasive ability. These data suggest new opportunities for diagnosis and treatment of metastatic melanoma.


Pigment Cell & Melanoma Research | 2017

Hypoxia-induced HIF1α targets in melanocytes reveal a molecular profile associated with poor melanoma prognosis

Stacie K. Loftus; Laura L. Baxter; Julia C. Cronin; Temesgen D. Fufa; William J. Pavan

Hypoxia and HIF1α signaling direct tissue‐specific gene responses regulating tumor progression, invasion, and metastasis. By integrating HIF1α knockdown and hypoxia‐induced gene expression changes, this study identifies a melanocyte‐specific, HIF1α‐dependent/hypoxia‐responsive gene expression signature. Integration of these gene expression changes with HIF1α ChIP‐Seq analysis identifies 81 HIF1α direct target genes in melanocytes. The expression levels for 10 of the HIF1α direct targets – GAPDH, PKM, PPAT, DARS, DTWD1, SEH1L, ZNF292, RLF, AGTRAP, and GPC6 – are significantly correlated with reduced time of disease‐free status in melanoma by logistic regression (P‐value = 0.0013) and ROC curve analysis (AUC = 0.826, P‐value < 0.0001). This HIF1α‐regulated profile defines a melanocyte‐specific response under hypoxia, and demonstrates the role of HIF1α as an invasive cell state gatekeeper in regulating cellular metabolism, chromatin and transcriptional regulation, vascularization, and invasion.

Collaboration


Dive into the Julia C. Cronin's collaboration.

Top Co-Authors

Avatar

Stacie K. Loftus

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William J. Pavan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yardena Samuels

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Steven A. Rosenberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John R. Wunderlich

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Neena S Agrawal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Todd D. Prickett

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xiaomu Wei

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jimmy Lin

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge