Network


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

Hotspot


Dive into the research topics where Mark P. Hamilton is active.

Publication


Featured researches published by Mark P. Hamilton.


Nature Communications | 2013

Identification of a pan-cancer oncogenic microRNA superfamily anchored by a central core seed motif

Mark P. Hamilton; Kimal Rajapakshe; Sean M. Hartig; Boris Reva; Michael D. McLellan; Cyriac Kandoth; Li Ding; Travis I. Zack; Preethi H. Gunaratne; David A. Wheeler; Cristian Coarfa; Sean E. McGuire

MicroRNAs modulate tumorigenesis through suppression of specific genes. As many tumour types rely on overlapping oncogenic pathways, a core set of microRNAs may exist, which consistently drives or suppresses tumorigenesis in many cancer types. Here we integrate The Cancer Genome Atlas (TCGA) pan-cancer data set with a microRNA target atlas composed of publicly available Argonaute Crosslinking Immunoprecipitation (AGO-CLIP) data to identify pan-tumour microRNA drivers of cancer. Through this analysis, we show a pan-cancer, coregulated oncogenic microRNA ‘superfamily’ consisting of the miR-17, miR-19, miR-130, miR-93, miR-18, miR-455 and miR-210 seed families, which cotargets critical tumour suppressors via a central GUGC core motif. We subsequently define mutations in microRNA target sites using the AGO-CLIP microRNA target atlas and TCGA exome-sequencing data. These combined analyses identify pan-cancer oncogenic cotargeting of the phosphoinositide 3-kinase, TGFβ and p53 pathways by the miR-17-19-130 superfamily members.


Trends in Endocrinology and Metabolism | 2015

The miRNA Interactome in Metabolic Homeostasis.

Sean M. Hartig; Mark P. Hamilton; David A. Bader; Sean E. McGuire

Global expression analyses demonstrate that alterations in miRNA levels correlate with various metabolic diseases. miRNAs regulate central metabolic pathways and thus play vital roles in maintaining organismal energy balance and metabolic homeostasis. Here we highlight novel sequencing technologies used to comprehensively define the target spectrum of miRNAs in metabolic disease that complement recent literature reporting physiologic roles for miRNAs in the regulation of glucose and lipid metabolism in peripheral tissues of animal models of metabolic dysfunction. These emerging technologies help decipher the complexity of the miRNA interactome and enrich our understanding of how miRNAs mediate physiologic effects by targeting a spectrum of gene transcripts simultaneously. miRNA-based therapeutics emerge as a viable strategy for treating metabolic diseases.


Cancer Cell | 2014

Copy Number Gain of hsa-miR-569 at 3q26.2 Leads to Loss of TP53INP1 and Aggressiveness of Epithelial Cancers

Pradeep Chaluvally-Raghavan; Fan Zhang; Sunila Pradeep; Mark P. Hamilton; Xi Zhao; Rajesha Rupaimoole; Tyler Moss; Yiling Lu; Shuangxing Yu; Chad V. Pecot; Miriam Ragle Aure; Sylvain Peuget; Cristian Rodriguez-Aguayo; Hee Dong Han; Dong Zhang; Avinashnarayan Venkatanarayan; Marit Krohn; Vessela N. Kristensen; Mihai Gagea; Prahlad T. Ram; Wenbin Liu; Gabriel Lopez-Berestein; Philip L. Lorenzi; Anne Lise Børresen-Dale; Koei Chin; Joe W. Gray; Nelson Dusetti; Sean E. McGuire; Elsa R. Flores; Anil K. Sood

Small noncoding miRNAs represent underexplored targets of genomic aberrations and emerging therapeutic targets. The 3q26.2 amplicon is among the most frequent genomic aberrations in multiple cancer lineages including ovarian and breast cancers. We demonstrate that hsa-miR-569 (hereafter designated as miR569), which is overexpressed in a subset of ovarian and breast cancers, at least in part due to the 3q26.2 amplicon, alters cell survival and proliferation. Downregulation of TP53INP1 expression by miR569 is required for the effects of miR569 on survival and proliferation. Targeting miR569 sensitizes ovarian and breast cancer cells overexpressing miR569 to cisplatin by increasing cell death both in vitro and in vivo. Thus targeting miR569 could potentially benefit patients with the 3q26.2 amplicon and subsequent miR569 elevation.


Molecular Oncology | 2016

SNHG16 is regulated by the Wnt pathway in colorectal cancer and affects genes involved in lipid metabolism

Lise Lotte Christensen; Kirsten True; Mark P. Hamilton; Morten Muhlig Nielsen; Nkerorema Djodji Damas; Christian Kroun Damgaard; Halit Ongen; Emmanouil T. Dermitzakis; Jesper B. Bramsen; Jakob Skou Pedersen; Anders H. Lund; Søren Vang; Katrine Stribolt; Mogens Rørbæk Madsen; Søren Laurberg; Sean E. McGuire; Torben F. Ørntoft; Claus L. Andersen

It is well established that lncRNAs are aberrantly expressed in cancer where they have been shown to act as oncogenes or tumor suppressors. RNA profiling of 314 colorectal adenomas/adenocarcinomas and 292 adjacent normal colon mucosa samples using RNA‐sequencing demonstrated that the snoRNA host gene 16 (SNHG16) is significantly up‐regulated in adenomas and all stages of CRC. SNHG16 expression was positively correlated to the expression of Wnt‐regulated transcription factors, including ASCL2, ETS2, and c‐Myc. In vitro abrogation of Wnt signaling in CRC cells reduced the expression of SNHG16 indicating that SNHG16 is regulated by the Wnt pathway. Silencing of SNHG16 resulted in reduced viability, increased apoptotic cell death and impaired cell migration. The SNHG16 silencing particularly affected expression of genes involved in lipid metabolism. A connection between SNHG16 and genes involved in lipid metabolism was also observed in clinical tumors. Argonaute CrossLinking and ImmunoPrecipitation (AGO‐CLIP) demonstrated that SNHG16 heavily binds AGO and has 27 AGO/miRNA target sites along its length, indicating that SNHG16 may act as a competing endogenous RNA (ceRNA) “sponging” miRNAs off their cognate targets. Most interestingly, half of the miRNA families with high confidence targets on SNHG16 also target the 3′UTR of Stearoyl‐CoA Desaturase (SCD). SCD is involved in lipid metabolism and is down‐regulated upon SNHG16 silencing. In conclusion, up‐regulation of SNHG16 is a frequent event in CRC, likely caused by deregulated Wnt signaling. In vitro analyses demonstrate that SNHG16 may play an oncogenic role in CRC and that it affects genes involved in lipid metabolism, possible through ceRNA related mechanisms.


Investigative Ophthalmology & Visual Science | 2014

Protein kinase C inhibitors sensitize GNAQ mutant uveal melanoma cells to ionizing radiation

Jasmina Z. Cerne; Sean M. Hartig; Mark P. Hamilton; Sue Anne Chew; Nicholas Mitsiades; Vassiliki Poulaki; Sean E. McGuire

PURPOSE Uveal melanoma (UM) tumors require large doses of radiation therapy (RT) to achieve tumor ablation, which frequently results in damage to adjacent normal tissues, leading to vision-threatening complications. Approximately 50% of UM patients present with activating somatic mutations in the gene encoding for G protein αq-subunit (GNAQ), which lead to constitutive activation of downstream pathways, including protein kinase C (PKC). In this study, we investigated the impact of small-molecule PKC inhibitors bisindolylmaleimide I (BIM) and sotrastaurin (AEB071), combined with ionizing radiation (IR), on survival in melanoma cell lines. METHODS Cellular radiosensitivity was determined by using a combination of proliferation, viability, and clonogenic assays. Cell-cycle effects were measured by flow cytometry. Transcriptomic and proteomic profiling were performed by quantitative real-time PCR, reverse-phase protein array analysis, and immunofluorescence. RESULTS We found that the PKC inhibitors combined with IR significantly decreased the viability, proliferation, and clonogenic potential of GNAQ(mt), but not GNAQ(wt)/BRAF(mt) cells, compared with IR alone. Combined treatment increased the antiproliferative and proapoptotic effects of IR in GNAQ(mt) cells through delayed DNA-damage resolution and enhanced induction of proteins involved in cell-cycle arrest, cell-growth arrest, and apoptosis. CONCLUSIONS Our preclinical results suggest that combined modality treatment may allow for reductions in the total RT dose and/or fraction size, which may lead to better functional organ preservation in the treatment of primary GNAQ(mt) UM. These findings suggest future clinical trials combining PKC inhibitors with RT in GNAQ(mt) UM warrant consideration.


Neoplasia | 2016

The Landscape of microRNA Targeting in Prostate Cancer Defined by AGO-PAR-CLIP.

Mark P. Hamilton; Kimal Rajapakshe; David A. Bader; Jasmina Z. Cerne; Eric A. Smith; Cristian Coarfa; Sean M. Hartig; Sean E. McGuire

MicroRNA (miRNA) deregulation in prostate cancer (PCa) contributes to PCa initiation and metastatic progression. To comprehensively define the cancer-associated changes in miRNA targeting and function in commonly studied models of PCa, we performed photoactivatable ribonucleoside-enhanced cross-linking immunoprecipitation of the Argonaute protein in a panel of PCa cell lines modeling different stages of PCa progression. Using this comprehensive catalogue of miRNA targets, we analyzed miRNA targeting on known drivers of PCa and examined tissue-specific and stage-specific pathway targeting by miRNAs. We found that androgen receptor is the most frequently targeted PCa oncogene and that miR-148a targets the largest number of known PCa drivers. Globally, tissue-specific and stage-specific changes in miRNA targeting are driven by homeostatic response to active oncogenic pathways. Our findings indicate that, even in advanced PCa, the miRNA pool adapts to regulate continuing alterations in the cancer genome to balance oncogenic molecular changes. These findings are important because they are the first to globally characterize miRNA changes in PCa and demonstrate how the miRNA target spectrum responds to staged tumorigenesis.


npj Genomic Medicine | 2017

Circular RNA expression is abundant and correlated to aggressiveness in early-stage bladder cancer

Trine Line Hauge Okholm; Morten Muhlig Nielsen; Mark P. Hamilton; Lise-Lotte Christensen; Søren Vang; Jakob Hedegaard; Thomas B. Hansen; Jørgen Kjems; Lars Dyrskjøt; Jakob Skou Pedersen

The functions and biomarker potential of circular RNAs (circRNAs) in various cancer types are a rising field of study, as emerging evidence relates circRNAs to tumorigenesis. Here, we profiled the expression of circRNAs in 457 tumors from patients with non-muscle-invasive bladder cancer (NMIBC). We show that a set of highly expressed circRNAs have conserved core splice sites, are associated with Alu repeats, and enriched with Synonymous Constraint Elements as well as microRNA target sites. We identified 113 abundant circRNAs that are differentially expressed between high and low-risk tumor subtypes. Analysis of progression-free survival revealed 13 circRNAs, among them circHIPK3 and circCDYL, where expression correlated with progression independently of the linear transcript and the host gene. In summary, our results demonstrate that abundant circRNAs possess multiple biological features, distinguishing them from low-expressed circRNAs and non-circularized exons, and suggest that circRNAs might serve as a new class of prognostic biomarkers in NMIBC.Bladder cancer: Circular RNAs show prognostic biomarker potentialExpression levels of non-coding “circular” RNA molecules could be used as a prognostic biomarker for patients with early-stage bladder cancer. A team led by Trine Line Hauge Okholm and Jakob Skou Pedersen from Aarhus University Hospital, Denmark, profiled the expression of these loop-forming, potentially gene-regulating RNAs in biopsied tumor samples from 457 patients with bladder cancer that had not invaded nearby muscle tissue. They identified a suite of 113 circular RNAs that were abundant and differentially expressed between patients with different molecular subtypes of bladder cancer. The researchers also found a smaller set of 13 circular RNAs for which expression levels correlated with disease progression. These non-coding RNA molecules, by indicating likely patient outcomes, could potentially serve as future diagnostic aids to inform treatment strategies and decisions.


Journal of Biological Chemistry | 2016

Mitochondrial activity in human white adipocytes is regulated by the ubiquitin carrier protein 9/microRNA-30a axis

Eun Hee Koh; Yong Chen; David A. Bader; Mark P. Hamilton; Bin He; Brian York; Shingo Kajimura; Sean E. McGuire; Sean M. Hartig

The acquisition of beige adipocyte features by white fat cells corresponds to protection against obesity-induced metabolic diseases in humans and animal models of type 2 diabetes. In adipose tissue, expression of the E2 small ubiquitin-like modifier ligase ubiquitin carrier protein 9 (Ubc9) is positively correlated with markers of insulin resistance and corresponds with impaired browning of human white adipocytes. However, the molecular regulation of Ubc9 expression in adipocytes and other cells remains unclear. In this study, we demonstrate that the mRNA and protein expression of Ubc9 are regulated by the microRNA miRNA-30a (miR-30a) in human subcutaneous adipocytes. Ubc9 and miR-30a exhibit inverse expression in adipose tissue, with miR-30a robustly elevated in brown fat. Depletion of Ubc9 by siRNA or enforced expression of a miR-30a mimic augments mitochondrial volume and respiration in human white adipocytes, reflecting features of brown fat cells. Furthermore, Ubc9 depletion induces a brown fat gene program in human subcutaneous adipocytes. Induction of the beige-selective gene program corresponds to stabilization of the PR domain-containing 16 (PRDM16) protein, an obligate transcriptional regulator of the brown/beige fat metabolic program in white adipocytes that interacts with Ubc9. Taken together, our data demonstrate a previously unappreciated molecular axis that controls browning of human white adipocytes.


Molecular Endocrinology | 2015

Ubc9 Impairs Activation of the Brown Fat Energy Metabolism Program in Human White Adipocytes

Sean M. Hartig; David A. Bader; Kathleen V. Abadie; Massoud Motamed; Mark P. Hamilton; Weiwen Long; Brian York; Michaela Mueller; Martin Wagner; Michael Trauner; Lawrence Chan; Mandeep Bajaj; David D. Moore; Michael A. Mancini; Sean E. McGuire

Insulin resistance and type 2 diabetes mellitus (T2DM) result from an inability to efficiently store and catabolize surplus energy in adipose tissue. Subcutaneous adipocytes protect against insulin resistance and T2DM by coupling differentiation with the induction of brown fat gene programs for efficient energy metabolism. Mechanisms that disrupt these programs in adipocytes are currently poorly defined, but represent therapeutic targets for the treatment of T2DM. To gain insight into these mechanisms, we performed a high-throughput microscopy screen that identified ubiquitin carrier protein 9 (Ubc9) as a negative regulator of energy storage in human sc adipocytes. Ubc9 depletion enhanced energy storage and induced the brown fat gene program in human sc adipocytes. Induction of adipocyte differentiation resulted in decreased Ubc9 expression commensurate with increased brown fat gene expression. Thiazolidinedione treatment reduced the interaction between Ubc9 and peroxisome proliferator-activated receptor (PPAR)γ, suggesting a mechanism by which Ubc9 represses PPARγ activity. In support of this hypothesis, Ubc9 overexpression remodeled energy metabolism in human sc adipocytes by selectively inhibiting brown adipocyte-specific function. Further, Ubc9 overexpression decreased uncoupling protein 1 expression by disrupting PPARγ binding at a critical uncoupling protein 1 enhancer region. Last, Ubc9 is significantly elevated in sc adipose tissue isolated from mouse models of insulin resistance as well as diabetic and insulin-resistant humans. Taken together, our findings demonstrate a critical role for Ubc9 in the regulation of sc adipocyte energy homeostasis.


Diabetes | 2018

miR-30a Remodels Subcutaneous Adipose Tissue Inflammation to Improve Insulin Sensitivity in Obesity

Eun-Hee Koh; Natasha Chernis; Pradip K. Saha; Liuling Xiao; David A. Bader; Bokai Zhu; Kimal Rajapakshe; Mark P. Hamilton; Xia Liu; Dimuthu Perera; Xi Chen; Brian York; Michael Trauner; Cristian Coarfa; Mandeep Bajaj; David D. Moore; Tuo Deng; Sean E. McGuire; Sean M. Hartig

Chronic inflammation accompanies obesity and limits subcutaneous white adipose tissue (WAT) expandability, accelerating the development of insulin resistance and type 2 diabetes mellitus. MicroRNAs (miRNAs) influence expression of many metabolic genes in fat cells, but physiological roles in WAT remain poorly characterized. Here, we report that expression of the miRNA miR-30a in subcutaneous WAT corresponds with insulin sensitivity in obese mice and humans. To examine the hypothesis that restoration of miR-30a expression in WAT improves insulin sensitivity, we injected adenovirus (Adv) expressing miR-30a into the subcutaneous fat pad of diabetic mice. Exogenous miR-30a expression in the subcutaneous WAT depot of obese mice coupled improved insulin sensitivity and increased energy expenditure with decreased ectopic fat deposition in the liver and reduced WAT inflammation. High-throughput proteomic profiling and RNA-Seq suggested that miR-30a targets the transcription factor STAT1 to limit the actions of the proinflammatory cytokine interferon-γ (IFN-γ) that would otherwise restrict WAT expansion and decrease insulin sensitivity. We further demonstrated that miR-30a opposes the actions of IFN-γ, suggesting an important role for miR-30a in defending adipocytes against proinflammatory cytokines that reduce peripheral insulin sensitivity. Together, our data identify a critical molecular signaling axis, elements of which are involved in uncoupling obesity from metabolic dysfunction.

Collaboration


Dive into the Mark P. Hamilton's collaboration.

Top Co-Authors

Avatar

Sean E. McGuire

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sean M. Hartig

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

David A. Bader

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Brian York

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Cristian Coarfa

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jasmina Z. Cerne

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kimal Rajapakshe

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

David D. Moore

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kjersti Aagaard

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Mandeep Bajaj

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge