Network


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

Hotspot


Dive into the research topics where Steven E. Robinson is active.

Publication


Featured researches published by Steven E. Robinson.


Nature Genetics | 2011

Exome sequencing identifies GRIN2A as frequently mutated in melanoma

Xiaomu Wei; Vijay Walia; Jimmy Lin; Jamie K. Teer; Todd D. Prickett; Jared J. Gartner; Sean Davis; Katherine Stemke-Hale; Michael A. Davies; Jeffrey E. Gershenwald; William H. Robinson; Steven E. Robinson; Steven A. Rosenberg; Yardena Samuels

The incidence of melanoma is increasing more than any other cancer, and knowledge of its genetic alterations is limited. To systematically analyze such alterations, we performed whole-exome sequencing of 14 matched normal and metastatic tumor DNAs. Using stringent criteria, we identified 68 genes that appeared to be somatically mutated at elevated frequency, many of which are not known to be genetically altered in tumors. Most importantly, we discovered that TRRAP harbored a recurrent mutation that clustered in one position (p. Ser722Phe) in 6 out of 167 affected individuals (∼4%), as well as a previously unidentified gene, GRIN2A, which was mutated in 33% of melanoma samples. The nature, pattern and functional evaluation of the TRRAP recurrent mutation suggest that TRRAP functions as an oncogene. Our study provides, to our knowledge, the most comprehensive map of genetic alterations in melanoma to date and suggests that the glutamate signaling pathway is involved in this disease.


Cancer Research | 2008

MicroRNA-137 Targets Microphthalmia-Associated Transcription Factor in Melanoma Cell Lines

Lynne T. Bemis; Robert W. Chen; Carol M. Amato; Elizabeth H. Classen; Steven E. Robinson; David Coffey; Paul F. Erickson; Yiqun G. Shellman; William A. Robinson

Micropthalmia-associated transcription factor (MITF) is the master regulator of melanocyte development, survival, and function. Frequent alteration in the expression of MITF is detected in melanoma, but the mechanism(s) underlying the alteration in expression have not been completely determined. In these studies, we have identified microRNA-137 (miR-137) as a regulator of MITF expression. The genomic locus of miR-137 at chromosome 1p22 places it in a region of the human genome previously determined to harbor an allele for melanoma susceptibility. Here, we show that expression of mature miR-137 in melanoma cell lines down-regulates MITF expression. Further, we have identified a 15-bp variable nucleotide tandem repeat located just 5 to the pre-miR-137 sequence, which alters the processing and function of miR-137 in melanoma cell lines.


Stem Cells | 2012

ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets.

Yuchun Luo; Katiuscia Dallaglio; Ying Chen; William A. Robinson; Steven E. Robinson; Martin D. McCarter; Jianbin Wang; Rene Gonzalez; David C. Thompson; David A. Norris; Dennis R. Roop; Vasilis Vasiliou; Mayumi Fujita

Although the concept of cancer stem cells (CSCs) is well‐accepted for many tumors, the existence of such cells in human melanoma has been the subject of debate. In this study, we demonstrate the existence of human melanoma cells that fulfill the criteria for CSCs (self‐renewal and differentiation) by serially xenotransplanting cells into nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. These cells possess high aldehyde dehydrogenase (ALDH) activity with ALDH1A1 and ALDH1A3 being the predominant ALDH isozymes. ALDH‐positive melanoma cells are more tumorigenic than ALDH‐negative cells in both NOD/SCID mice and NSG mice. Biological analyses of the ALDH‐positive melanoma cells reveal the ALDH isozymes to be key molecules regulating the function of these cells. Silencing ALDH1A by siRNA or shRNA leads to cell cycle arrest, apoptosis, decreased cell viability in vitro, and reduced tumorigenesis in vivo. ALDH‐positive melanoma cells are more resistant to chemotherapeutic agents and silencing ALDH1A by siRNA sensitizes melanoma cells to drug‐induced cell death. Furthermore, we, for the first time, examined the molecular signatures of ALDH‐positive CSCs from patient‐derived tumor specimens. The signatures of melanoma CSCs include retinoic acid (RA)‐driven target genes with RA response elements and genes associated with stem cell function. These findings implicate that ALDH isozymes are not only biomarkers of CSCs but also attractive therapeutic targets for human melanoma. Further investigation of these isozymes and genes will enhance our understanding of the molecular mechanisms governing CSCs and reveal new molecular targets for therapeutic intervention of cancer. STEM Cells2012;30:2100–2113


Proceedings of the National Academy of Sciences of the United States of America | 2013

Whole-genome sequencing identifies a recurrent functional synonymous mutation in melanoma

Jared J. Gartner; Stephen C. J. Parker; Todd D. Prickett; Ken Dutton-Regester; Michael L. Stitzel; Jimmy C. Lin; Sean Davis; Vijaya L. Simhadri; Sujata Jha; Nobuko Katagiri; Valer Gotea; Jamie K. Teer; Xiaomu Wei; Mario A. Morken; Umesh Bhanot; Guo Chen; Laura Elnitski; Michael A. Davies; Jeffrey E. Gershenwald; Hannah Carter; Rachel Karchin; William H. Robinson; Steven E. Robinson; Steven A. Rosenberg; Francis S. Collins; Giovanni Parmigiani; Anton A. Komar; Chava Kimchi-Sarfaty; Nicholas K. Hayward; Elliott H. Margulies

Synonymous mutations, which do not alter the protein sequence, have been shown to affect protein function [Sauna ZE, Kimchi-Sarfaty C (2011) Nat Rev Genet 12(10):683–691]. However, synonymous mutations are rarely investigated in the cancer genomics field. We used whole-genome and -exome sequencing to identify somatic mutations in 29 melanoma samples. Validation of one synonymous somatic mutation in BCL2L12 in 285 samples identified 12 cases that harbored the recurrent F17F mutation. This mutation led to increased BCL2L12 mRNA and protein levels because of differential targeting of WT and mutant BCL2L12 by hsa-miR-671–5p. Protein made from mutant BCL2L12 transcript bound p53, inhibited UV-induced apoptosis more efficiently than WT BCL2L12, and reduced endogenous p53 target gene transcription. This report shows selection of a recurrent somatic synonymous mutation in cancer. Our data indicate that silent alterations have a role to play in human cancer, emphasizing the importance of their investigation in future cancer genome studies.


Journal of Investigative Dermatology | 2012

Side Population Cells from Human Melanoma Tumors Reveal Diverse Mechanisms for Chemoresistance

Yuchun Luo; Lixia Z. Ellis; Katiuscia Dallaglio; Moe Takeda; William A. Robinson; Steven E. Robinson; Weimin Liu; Karl D. Lewis; Martin D. McCarter; Rene Gonzalez; David A. Norris; Dennis R. Roop; Richard A. Spritz; Natalie G. Ahn; Mayumi Fujita

Side population (SP) is identified as cells capable of excluding the fluorescent Hoechst dye and anticancer drugs, and represents hematopoietic stem cells and chemoresistant cells from several solid tumors. In this study, we confirmed the presence of SP cells in tumors from melanoma patients. Melanoma SP cells overexpressed ATP-binding-cassette (ABC) transporters, ABCB1 and ABCB5. We generated a direct in vivo xenograft model, and demonstrated that SP cells were resistant to paclitaxel, a substrate of ABCB1, both in vitro and in vivo. However, melanoma SP cells were also resistant to temozolomide, which is not a substrate for ABC transporters, through IL-8 upregulation. In addition, gene profiling studies identified three signaling pathways (NF- κB, α6-β4-integrin and IL-1) as differentially upregulated in melanoma SP cells, and there was a significant increase of PCDHB11 and decrease of FUK and TBX2 in these cells. Therefore, we provide evidence that SP is an enriched source of chemoresistant cells in human melanomas, and suggest that the selected genes and signaling pathways of SP cells may be a potential target for effective melanoma therapies. To our knowledge, this is previously unreported study to isolate SP cells from melanoma patients and to investigate the gene expression profiling of these cells.


Pigment Cell & Melanoma Research | 2010

p53 prevents progression of nevi to melanoma predominantly through cell cycle regulation

Tamara Terzian; Enrique C. Torchia; Daisy Dai; Steven E. Robinson; Kazutoshi Murao; Regan A. Stiegmann; Victoria Gonzalez; Glen M. Boyle; Marianne Broome Powell; Pamela M. Pollock; Guillermina Lozano; William A. Robinson; Dennis R. Roop; Neil F. Box

p53 is the central member of a critical tumor suppressor pathway in virtually all tumor types, where it is silenced mainly by missense mutations. In melanoma, p53 predominantly remains wild type, thus its role has been neglected. To study the effect of p53 on melanocyte function and melanomagenesis, we crossed the ‘high‐p53’Mdm4+/− mouse to the well‐established TP‐ras0/+ murine melanoma progression model. After treatment with the carcinogen dimethylbenzanthracene (DMBA), TP‐ras0/+ mice on the Mdm4+/− background developed fewer tumors with a delay in the age of onset of melanomas compared to TP‐ras0/+ mice. Furthermore, we observed a dramatic decrease in tumor growth, lack of metastasis with increased survival of TP‐ras0/+: Mdm4+/− mice. Thus, p53 effectively prevented the conversion of small benign tumors to malignant and metastatic melanoma. p53 activation in cultured primary melanocyte and melanoma cell lines using Nutlin‐3, a specific Mdm2 antagonist, supported these findings. Moreover, global gene expression and network analysis of Nutlin‐3‐treated primary human melanocytes indicated that cell cycle regulation through the p21WAF1/CIP1 signaling network may be the key anti‐melanomagenic activity of p53.


Journal of Investigative Dermatology | 2015

Narrow Band Ultraviolet B Treatment for Human Vitiligo Is Associated with Proliferation, Migration, and Differentiation of Melanocyte Precursors

Nathaniel B. Goldstein; Maranke I. Koster; Laura G. Hoaglin; Nicole S. Spoelstra; Katerina Kechris; Steven E. Robinson; William A. Robinson; Dennis R. Roop; David A. Norris; Stanca A. Birlea

In vitiligo, the autoimmune destruction of epidermal melanocytes produces white spots that can be repigmented by melanocyte precursors from the hair follicles, following stimulation with UV light. We examined by immunofluorescence the distribution of melanocyte markers (C-KIT, DCT, PAX3, and TYR) coupled with markers of proliferation (KI-67) and migration (MCAM) in precursors and mature melanocytes from the hair follicle and the epidermis of untreated and narrow band UVB (NBUVB)-treated human vitiligo skin. NBUVB was associated with a significant increase in the number of melanocytes in the infundibulum and with restoration of the normal melanocyte population in the epidermis, which was lacking in the untreated vitiligo. We identified several precursor populations (melanocyte stem cells, melanoblasts, and other immature phenotypes), and progressively differentiating melanocytes, some with putative migratory and/or proliferative abilities. The primary melanocyte germ was present in the untreated and treated hair follicle bulge, whereas a possible secondary melanocyte germ composed of C-KIT+ melanocytes was found in the infundibulum and interfollicular epidermis of UV-treated vitiligo. This is an exceptional model for studying the mobilization of melanocyte stem cells in human skin. Improved understanding of this process is essential for designing better treatments for vitiligo, ultimately based on melanocyte stem cell activation and mobilization.


PLOS ONE | 2013

Extracellular Vesicles Secreted from Cancer Cell Lines Stimulate Secretion of MMP-9, IL-6, TGF-β1 and EMMPRIN

Jasmina S. Redzic; Agnieszka A. Kendrick; Karim Bahmed; Kristin D. Dahl; Chad G. Pearson; William A. Robinson; Steven E. Robinson; Michael W. Graner; Elan Z. Eisenmesser

Extracellular vesicles (EVs) are key contributors to cancer where they play an integral role in cell-cell communication and transfer pro-oncogenic molecules to recipient cells thereby conferring a cancerous phenotype. Here, we purified EVs using straightforward biochemical approaches from multiple cancer cell lines and subsequently characterized these EVs via multiple biochemical and biophysical methods. In addition, we used fluorescence microscopy to directly show internalization of EVs into the recipient cells within a few minutes upon addition of EVs to recipient cells. We confirmed that the transmembrane protein EMMPRIN, postulated to be a marker of EVs, was indeed secreted from all cell lines studied here. We evaluated the response to EV stimulation in several different types of recipient cells lines and measured the ability of these purified EVs to induce secretion of several factors highly upregulated in human cancers. Our data indicate that purified EVs preferentially stimulate secretion of several proteins implicated in driving cancer in monocytic cells but only harbor limited activity in epithelial cells. Specifically, we show that EVs are potent stimulators of MMP-9, IL-6, TGF-β1 and induce the secretion of extracellular EMMPRIN, which all play a role in driving immune evasion, invasion and inflammation in the tumor microenvironment. Thus, by using a comprehensive approach that includes biochemical, biological, and spectroscopic methods, we have begun to elucidate the stimulatory roles.


PLOS ONE | 2011

Transcriptome Profiling of Whole Blood Cells Identifies PLEK2 and C1QB in Human Melanoma

Yuchun Luo; Steven E. Robinson; Junichi Fujita; Lisa Siconolfi; Jay Magidson; Carl K. Edwards; Karl Wassmann; Kathleen Storm; David A. Norris; Danute Bankaitis-Davis; William A. Robinson; Mayumi Fujita

Background Developing analytical methodologies to identify biomarkers in easily accessible body fluids is highly valuable for the early diagnosis and management of cancer patients. Peripheral whole blood is a “nucleic acid-rich” and “inflammatory cell-rich” information reservoir and represents systemic processes altered by the presence of cancer cells. Methodology/Principal Findings We conducted transcriptome profiling of whole blood cells from melanoma patients. To overcome challenges associated with blood-based transcriptome analysis, we used a PAXgene™ tube and NuGEN Ovation™ globin reduction system. The combined use of these systems in microarray resulted in the identification of 78 unique genes differentially expressed in the blood of melanoma patients. Of these, 68 genes were further analyzed by quantitative reverse transcriptase PCR using blood samples from 45 newly diagnosed melanoma patients (stage I to IV) and 50 healthy control individuals. Thirty-nine genes were verified to be differentially expressed in blood samples from melanoma patients. A stepwise logit analysis selected eighteen 2-gene signatures that distinguish melanoma from healthy controls. Of these, a 2-gene signature consisting of PLEK2 and C1QB led to the best result that correctly classified 93.3% melanoma patients and 90% healthy controls. Both genes were upregulated in blood samples of melanoma patients from all stages. Further analysis using blood fractionation showed that CD45− and CD45+ populations were responsible for the altered expression levels of PLEK2 and C1QB, respectively. Conclusions/Significance The current study provides the first analysis of whole blood-based transcriptome biomarkers for malignant melanoma. The expression of PLEK2, the strongest gene to classify melanoma patients, in CD45− subsets illustrates the importance of analyzing whole blood cells for biomarker studies. The study suggests that transcriptome profiling of blood cells could be used for both early detection of melanoma and monitoring of patients for residual disease.


Journal of Investigative Dermatology | 2015

Combining a BCL2 Inhibitor with the Retinoid Derivative Fenretinide Targets Melanoma Cells Including Melanoma Initiating Cells

Nabanita Mukherjee; Steven N. Reuland; Yan Lu; Yuchun Luo; Karoline Lambert; Mayumi Fujita; William A. Robinson; Steven E. Robinson; David A. Norris; Yiqun G. Shellman

Investigations from multiple laboratories support the existence of melanoma initiating cells (MICs) that potentially contribute to melanomas drug resistance. ABT-737, a small molecule BCL-2/BCL-XL/BCL-W inhibitor, is promising in cancer treatments, but not very effective against melanoma, with the anti-apoptotic protein MCL-1 as the main contributor to resistance. The synthetic retinoid fenretinide (4-HPR) has shown promise for treating breast cancers. Here, we tested whether the combination of ABT-737 with 4-HPR is effective in killing both the bulk of melanoma cells and MICs. The combination synergistically decreased cell viability and caused cell death in multiple melanoma cells lines (carrying either BRAF or NRAS mutations), but not in normal melanocytes. The combination increased the NOXA expression and caspase-dependent MCL-1 degradation. Knocking-down NOXA protected cells from combination-induced apoptosis, implicating the role of NOXA in the drug synergy. The combination treatment also disrupted primary spheres (a functional assay for MICs) and decreased the percentage of ALDHhigh cells (a marker of MICs) in melanoma cell lines. Moreover, the combination inhibited the self-renewal capacity of MICs, measured by secondary sphere forming assays. In vivo, the combination inhibited tumor growth. Thus, this combination is a promising treatment strategy for melanoma, regardless of mutation status of BRAF or NRAS.

Collaboration


Dive into the Steven E. Robinson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Norris

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Carol M. Amato

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Yiqun G. Shellman

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dennis R. Roop

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Mayumi Fujita

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Yuchun Luo

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge