Network


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

Hotspot


Dive into the research topics where Anne E. Sarver is active.

Publication


Featured researches published by Anne E. Sarver.


PLOS ONE | 2016

MicroRNA Mediated Chemokine Responses in Human Airway Smooth Muscle Cells

Mythili Dileepan; Anne E. Sarver; Savita P. Rao; Reynold A. Panettieri; Subbaya Subramanian; Mathur S. Kannan

Airway smooth muscle (ASM) cells play a critical role in the pathophysiology of asthma due to their hypercontractility and their ability to proliferate and secrete inflammatory mediators. microRNAs (miRNAs) are gene regulators that control many signaling pathways and thus serve as potential therapeutic alternatives for many diseases. We have previously shown that miR-708 and miR-140-3p regulate the MAPK and PI3K signaling pathways in human ASM (HASM) cells following TNF-α exposure. In this study, we investigated the regulatory effect of these miRNAs on other asthma-related genes. Microarray analysis using the Illumina platform was performed with total RNA extracted from miR-708 (or control miR)-transfected HASM cells. Inhibition of candidate inflammation-associated gene expression was further validated by qPCR and ELISA. The most significant biologic functions for the differentially expressed gene set included decreased inflammatory response, cytokine expression and signaling. qPCR revealed inhibition of expression of CCL11, CXCL10, CCL2 and CXCL8, while the release of CCL11 was inhibited in miR-708-transfected cells. Transfection of cells with miR-140-3p resulted in inhibition of expression of CCL11, CXCL12, CXCL10, CCL5 and CXCL8 and of TNF-α-induced CXCL12 release. In addition, expression of RARRES2, CD44 and ADAM33, genes known to contribute to the pathophysiology of asthma, were found to be inhibited in miR-708-transfected cells. These results demonstrate that miR-708 and miR-140-3p exert distinct effects on inflammation-associated gene expression and biological function of ASM cells. Targeting these miRNA networks may provide a novel therapeutic mechanism to down-regulate airway inflammation and ASM proliferation in asthma.


Oncotarget | 2016

Imprinting defects at human 14q32 locus alters gene expression and is associated with the pathobiology of osteosarcoma

Jingmin Shu; Lihua Li; Anne E. Sarver; Emily A. Pope; Jyotika Varshney; Venugopal Thayanithy; Logan G. Spector; David A. Largaespada; Clifford J. Steer; Subbaya Subramanian

Osteosarcoma is the most common primary bone malignancy affecting children and adolescents. Although several genetic predisposing conditions have been associated with osteosarcoma, our understanding of its pathobiology is rather limited. Here we show that, first, an imprinting defect at human 14q32-locus is highly prevalent (87%) and specifically associated with osteosarcoma patients < 30 years of age. Second, the average demethylation at differentially methylated regions (DMRs) in the 14q32-locus varied significantly compared to genome-wide demethylation. Third, the 14q32-locus was enriched in both H3K4-me3 and H3K27-me3 histone modifications that affected expression of all imprinted genes and miRNAs in this region. Fourth, imprinting defects at 14q32 - DMRs are present in triad DNA samples from affected children and their biological parents. Finally, imprinting defects at 14q32-DMRs were also observed at higher frequencies in an Rb1/Trp53 mutation-induced osteosarcoma mouse model. Further analysis of normal and tumor tissues from a Sleeping Beauty mouse model of spontaneous osteosarcoma supported the notion that these imprinting defects may be a key factor in osteosarcoma pathobiology. In conclusion, we demonstrate that imprinting defects at the 14q32 locus significantly alter gene expression, may contribute to the pathogenesis of osteosarcoma, and could be predictive of survival outcomes.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2016

Comprehensive analysis of microRNA signature of mouse pancreatic acini: overexpression of miR-21-3p in acute pancreatitis

Ajay Dixit; Anne E. Sarver; Zuobiao Yuan; John George; Usman Barlass; Hassam Cheema; Archana Sareen; Sulagna Banerjee; Vikas Dudeja; Rajinder Dawra; Subbaya Subramanian; Ashok K. Saluja

In the current study, we have characterized the global miRNA expression profile in mouse pancreatic acinar cells and during acute pancreatitis using next-generation RNA sequencing. We identified 324 known and six novel miRNAs that are expressed in mouse pancreatic acinar cells. In the basal state, miR-148a-3p, miR-375-3p, miR-217-5p, and miR-200a-3p were among the most abundantly expressed, whereas miR-24-5p and miR-421-3p were the least abundant. Treatment of acinar cells with caerulein (100 nM) and taurolithocholic acid 3-sulfate [TLC-S (250 μM)] induced numerous changes in miRNA expression profile. In particular, we found significant overexpression of miR-21-3p in acini treated with caerulein and TLC-S. We further looked at the expression of miR-21-3p in caerulein, l-arginine, and caerulein + LPS-induced acute pancreatitis mouse models and found 12-, 21-, and 50-fold increased expression in the pancreas, respectively. In summary, this is the first comprehensive analysis of global miRNA expression profile of mouse pancreatic acinar cells in normal and disease conditions. Our analysis shows that miR-21-3p expression level correlates with the severity of the disease.


Cancer Research | 2018

Comparative transcriptome analysis quantifies immune cell transcript levels, metastatic progression and survival in osteosarcoma

Milcah C. Scott; Nuri A. Temiz; Anne E. Sarver; Rebecca S. LaRue; Susan K. Rathe; Jyotika Varshney; Natalie K. Wolf; Branden S. Moriarity; Timothy D. O'Brien; Logan G. Spector; David A. Largaespada; Jaime F. Modiano; Subbaya Subramanian; Aaron L. Sarver

Overall survival of patients with osteosarcoma (OS) has improved little in the past three decades, and better models for study are needed. OS is common in large dog breeds and is genetically inducible in mice, making the disease ideal for comparative genomic analyses across species. Understanding the level of conservation of intertumor transcriptional variation across species and how it is associated with progression to metastasis will enable us to more efficiently develop effective strategies to manage OS and to improve therapy. In this study, transcriptional profiles of OS tumors and cell lines derived from humans (n = 49), mice (n = 103), and dogs (n = 34) were generated using RNA sequencing. Conserved intertumor transcriptional variation was present in tumor sets from all three species and comprised gene clusters associated with cell cycle and mitosis and with the presence or absence of immune cells. Further, we developed a novel gene cluster expression summary score (GCESS) to quantify intertumor transcriptional variation and demonstrated that these GCESS values associated with patient outcome. Human OS tumors with GCESS values suggesting decreased immune cell presence were associated with metastasis and poor survival. We validated these results in an independent human OS tumor cohort and in 15 different tumor data sets obtained from The Cancer Genome Atlas. Our results suggest that quantification of immune cell absence and tumor cell proliferation may better inform therapeutic decisions and improve overall survival for OS patients.Significance: This study offers new tools to quantify tumor heterogeneity in osteosarcoma, identifying potentially useful prognostic biomarkers for metastatic progression and survival in patients. Cancer Res; 78(2); 326-37. ©2017 AACR.


Laboratory Investigation | 2015

Identification, by systematic RNA sequencing, of novel candidate biomarkers and therapeutic targets in human soft tissue tumors

Anne E. Sarver; Aaron L. Sarver; Venugopal Thayanithy; Subbaya Subramanian

Human sarcomas comprise a heterogeneous group of more than 50 subtypes broadly classified into two groups: bone and soft tissue sarcomas. Such heterogeneity and their relative rarity have made them challenging targets for classification, biomarker identification, and development of improved treatment strategies. In this study, we used RNA sequencing to analyze 35 primary human tissue samples representing 13 different sarcoma subtypes, along with benign schwannoma, and normal bone and muscle tissues. For each sarcoma subtype, we detected unique messenger RNA (mRNA) expression signatures, which we further subjected to bioinformatic functional analysis, upstream regulatory analysis, and microRNA (miRNA) targeting analysis. We found that, for each sarcoma subtype, significantly upregulated genes and their deduced upstream regulators included not only previously implicated known players but also novel candidates not previously reported to be associated with sarcoma. For example, the schwannoma samples were characterized by high expression of not only the known associated proteins GFAP and GAP43 but also the novel player GJB6. Further, when we integrated our expression profiles with miRNA expression data from each sarcoma subtype, we were able to deduce potential key miRNA–gene regulator relationships for each. In the Ewing’s sarcoma and fibromatosis samples, two sarcomas where miR-182-5p is significantly downregulated, multiple predicted targets were significantly upregulated, including HMCN1, NKX2-2, SCNN1G, and SOX2. In conclusion, despite the small number of samples per sarcoma subtype, we were able to identify key known players; concurrently, we discovered novel genes that may prove to be important in the molecular classification of sarcomas and in the development of novel treatments.


Advances in Experimental Medicine and Biology | 2015

microRNAs in the Malignant Transformation Process

Anne E. Sarver; Lihua Li; Reena V. Kartha; Subbaya Subramanian

Many cancers originate as benign neoplasms that transform into malignant cancerous tumors in a multistep progression that is regulated, in part, by microRNAs. Benign neoplasms, by definition, lack the ability to invade adjacent tissues or spread to distant sites through metastasis. The benign to malignant transition is a critical intervention stage as tumors diagnosed in subsequent nonlocalized and malignant stages are exponentially more difficult to treat successfully. This chapter explores the critical roles that microRNAs play in the transformation from benign to malignant in four representative cancers: colorectal cancer, pancreatic cancer, malignant peripheral nerve sheath tumor, and prostate cancer. Understanding how these microRNAs control this progression and transformation will lead to new therapeutic targets and diagnostic biomarkers, resulting in improved treatments and patient outcomes.


bioRxiv | 2018

OMCD: OncoMir Cancer Database

Aaron L. Sarver; Anne E. Sarver; Ce Yuan; Subbaya Subramanian

MicroRNAs (miRNAs) are crucially important in the development of cancer. Their dysregulation, commonly observed in various types of cancer, is largely cancer-dependent. Thus, to understand the tumor biology and to develop accurate and sensitive biomarkers, we need to understand pan-cancer miRNA expression. At the University of Minnesota, we developed the OncoMir Cancer Database (OMCD), hosted on a web server, which allows easy and systematic comparative genomic analyses of miRNA sequencing data derived from more than 9,500 cancer patients tissue samples available in the Cancer Genome Atlas (TCGA). OMCD includes associated clinical information and is searchable by organ-specific terms common to the TCGA. Freely available to all users (www.oncomir.umn.edu/omcd/), OMCD enables (1) simple visualization of TCGA miRNA sequencing data, (2) statistical analysis of differentially expressed miRNAs for each cancer type, and (3) exploration of miRNA clusters across cancer types. Database URL www.oncomir.umn.edu/omcd


Epigenomics | 2016

Imprinting defects in osteosarcoma: DNA- and chromatin-modifying drugs hold promise for osteosarcoma therapy

Anne E. Sarver; Subbaya Subramanian

The overall survival rate of patients with metastatic osteosarcoma has not improved over past three decades. In more than 70% of patients, current standard-of-care therapies ultimately fail to prevent relapse and metastasis, indicating a critical need for novel drugs [1]. Osteosarcoma may be considered to be primarily a disease of development, with the majority of cases occurring in pediatric patients. Inherited factors play a greater role in pediatric tumorigenesis compared with cancers typically appearing later in life due to environmental exposure and/or serial accumulation of genetic mutations. Our laboratory discovered that children diagnosed with osteosarcoma may be impacted by a 14q32 locus DNA imprinting defect that was also found in the patient’s parents but not in unrelated healthy individuals that appears to result in dysregulation of a large number of miRNAs and genes that normally function as tumor suppressors [2]. Based on this we propose considering a complementary treatment strategy utilizing DNAand chromatin-modifying drugs that would shift the cellular balance from proliferation to differentiation, while also sensitizing these cells to existing standard-of-care therapies. Transcriptional regulation by DNA methylation was first recognized over 40 years ago and initially proposed as a mechanism for X-chromosome inactivation [3,4]. Interestingly, studies have shown that epigenetic signatures can change over time due to aging and may be accelerated by disease [5] or environmental factors such as diet [6]. Recent work suggests that up to 85% of the variance in mutation rates along cancer genomes is a result of replication timing, chromatin accessibility and modification [7]. Allele-specific expression is controlled in part by epigenetic DNA imprinting and some genes show distinct maternal and paternal allele methylation patterns. To answer the question of whether or not pathogenic DNA imprinting defects are present and detectable in the unaffected parents of pediatric osteosarcoma patients our laboratory, in collaboration, assessed DNA methylation changes and histone modifications in their tissue samples and found that an imprinting defect at the 14q32 locus was highly prevalent and specifically associated with osteosarcoma [2]. DNA methylation typically comprises the addition of methyl groups to a cluster of sites, usually CpG dinucleotides, in the promoter or enhancer regions of a gene functioning as an ‘off switch’ that blocks transcription. This methylation signature is maintained during cellular division by a family of cellular enzymes known as the DNMTs. DNMT activity is mediated by a family of evolutionarily conserved enzymes, ten-eleven translocation methylcytosine dioxygenases (TETs). These TET enzymes are dependent on α-ketoglutarate, a metabolite produced by isocitrate dehydrogenases (IDHs), and their activity is enhanced by their ascorbic Anne E Sarver Department of Surgery, University of Minnesota, 11–212 Moos Tower (Mail code: MMC 195) 420 Delaware St, S.E Minneapolis, MN 55455, USA


Cancer Research | 2016

Abstract 1097: The miR-17-92 microRNA cluster plays a crucial role in osteosarcoma progression

Jyotika Varshney; Nicholas J. Slipek; John Osborne; Adrian Chang; Anne E. Sarver; Ingrid Cornax; Gerry O`Sullivan; Subbaya Subramanian; David A. Largaespada

Osteosarcoma is the most common primary bone malignancy that affects adolescents. Around 30% of patients with localized osteosarcoma and 70% of patients with metastasis will experience treatment failure within 5 years of diagnosis. The complex biology of osteosarcoma and astounding genetic heterogeneity has made it challenging to identify effective new gene targets and therapeutic agents. Our studies found an overall overexpression of a microRNA cluster, miR-17-92 microRNAs in human primary osteosarcoma compared to normal bone. We learned that upregulation of this miR-17-92 cluster simultaneously silences a suite of key tumor suppressors. Using data from a novel spontaneous osteosarcoma mouse model and genetic screen, we discovered miR-17-92 targets, such as PTEN, PTRPD and SRGAP2, which are potential tumor suppressor genes. Specifically blocking miR-17-92 function in osteosarcoma cells reduced their migration and ability to grow larger tumors in immunodeficient mice compared to the controls. Also, knockdown of miR-17-92 cluster microRNAs led to increase in the levels of PTPRD and SRGAP2 in cells as well as tumors; further suggesting that miR-17-92 is targeting these genes. We also performed gain-of-function of miR-17-92 studies in a poorly aggressive osteosarcoma cell line and found that overexpression of miR-17-92 leads to ability to grow in an anchorage independent manner and form tumors in immunodeficient mice, both features that are lacking in the parental line. Ongoing RNA sequencing studies on miR-17-92 target transcripts in osteosarcoma cells, and functional analyses of miR-17-92 deletion mutants, will be presented. In an attempt to target miR-17-92 miRNA expression, we have tested small molecules. Our data suggests that triptolide, a diterpenoid epoxide, inhibits MYC expression and downregulates miR-17-92 miRNAs resulting in upregulation of several tumor suppressor driver proteins including PTEN, PTPRD, and SRGAP2. Together, our data suggests that upregulation of miR-17-92 miRNAs contributes to osteosarcoma progression and triptolide inhibits miR-17-92 expression. These data have implications for how sarcomas develop in general and suggest a new way to treat cancer by targeting microRNAs using small molecules. Citation Format: Jyotika Varshney, Nicholas J. Slipek, John Osborne, Adrian Chang, Anne E. Sarver, Ingrid Cornax, Gerry M. O’ Sullivan, Subbaya Subramanian, David A. Largaespada. The miR-17-92 microRNA cluster plays a crucial role in osteosarcoma progression. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1097.


Cancer Research | 2016

Abstract B24: Identification of immunosuppressive networks in colon cancer

Anne E. Sarver; Aaron L. Sarver; Subbaya Subramanian

Despite important improvements over the past two decades in preventive screening and targeted therapies, colorectal cancer remains the third major cause of cancer-related deaths in the U.S. The majority (~85%) of colon cancer tumors are non-immunogenic, i.e. they lack a significant number of infiltrating cytotoxic T cells, and are typically unresponsive to the new immune-checkpoint inhibitor based therapies that have dramatically changed the way we treat many cancer patients. These non-immunogenic colon cancer tumors are also characterized by chromosome instability and are microsatellite stable (MSS). Microsatellite instable (MSI) tumors present a greater variety of antigens and are significantly more immunogenic. We analyzed the paired mRNA and microRNA RNA-Seq expression profiles contained in The Cancer Genome Atlas (TCGA), representing MSS colon cancer, MSI colon cancer, and normal colon tissues. These expression profiles reflect the heterogeneity of the tumor samples, with MSI samples showing distinct immune cell signatures. Integrative analysis of mRNA and microRNA expression patterns revealed signaling networks that may represent, in part, the molecular mechanisms by which cancer cells modulate the host response to create an immune privileged microenvironment. These findings could result in a novel therapeutic avenue, targeting a tumor9s ability to suppress the immune response, addressing a critical barrier to treatment for non-immunogenic tumors. Citation Format: Anne E. Sarver, Aaron L. Sarver, Subbaya Subramanian. Identification of immunosuppressive networks in colon cancer. [abstract]. In: Proceedings of the AACR Special Conference on Noncoding RNAs and Cancer: Mechanisms to Medicines ; 2015 Dec 4-7; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2016;76(6 Suppl):Abstract nr B24.

Collaboration


Dive into the Anne E. Sarver's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge