Network


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

Hotspot


Dive into the research topics where Timothy L. Mosbruger is active.

Publication


Featured researches published by Timothy L. Mosbruger.


Nature Communications | 2015

Exosome-delivered microRNAs modulate the inflammatory response to endotoxin.

Margaret Alexander; Ruozhen Hu; Marah C. Runtsch; Dominique A. Kagele; Timothy L. Mosbruger; Tanya Tolmachova; Miguel C. Seabra; June L. Round; Diane M. Ward; Ryan M. O'Connell

MicroRNAs regulate gene expression posttranscriptionally and function within the cells in which they are transcribed. However, recent evidence suggests that microRNAs can be transferred between cells and mediate target gene repression. We find that endogenous miR-155 and miR-146a, two critical microRNAs that regulate inflammation, are released from dendritic cells within exosomes and are subsequently taken up by recipient dendritic cells. Following uptake, exogenous microRNAs mediate target gene repression and can reprogramme the cellular response to endotoxin, where exosome-delivered miR-155 enhances while miR-146a reduces inflammatory gene expression. We also find that miR-155 and miR-146a are present in exosomes and pass between immune cells in vivo, as well as demonstrate that exosomal miR-146a inhibits while miR-155 promotes endotoxin-induced inflammation in mice. Together, our findings provide strong evidence that endogenous microRNAs undergo a functional transfer between immune cells and constitute a mechanism of regulating the inflammatory response.


PLOS ONE | 2016

Genome-Wide CRISPR-Cas9 Screen Identifies MicroRNAs That Regulate Myeloid Leukemia Cell Growth

Jared Wallace; Ruozhen Hu; Timothy L. Mosbruger; Timothy J. Dahlem; W. Zac Stephens; Dinesh S. Rao; June L. Round; Ryan M. O’Connell

Mammalian microRNA expression is dysregulated in human cancer. However, the functional relevance of many microRNAs in the context of tumor biology remains unclear. Using CRISPR-Cas9 technology, we performed a global loss-of-function screen to simultaneously test the functions of individual microRNAs and protein-coding genes during the growth of a myeloid leukemia cell line. This approach identified evolutionarily conserved human microRNAs that suppress or promote cell growth, revealing that microRNAs are extensively integrated into the molecular networks that control tumor cell physiology. miR-155 was identified as a top microRNA candidate promoting cellular fitness, which we confirmed with two distinct miR-155-targeting CRISPR-Cas9 lentiviral constructs. Further, we performed anti-correlation functional profiling to predict relevant microRNA-tumor suppressor gene or microRNA-oncogene interactions in these cells. This analysis identified miR-150 targeting of p53, a connection that was experimentally validated. Taken together, our study describes a powerful genetic approach by which the function of individual microRNAs can be assessed on a global level, and its use will rapidly advance our understanding of how microRNAs contribute to human disease.


American Journal of Clinical Pathology | 2014

A Comparison of Deep Sequencing of TCRG Rearrangements vs Traditional Capillary Electrophoresis for Assessment of Clonality in T-Cell Lymphoproliferative Disorders

Jonathan A. Schumacher; Eric J. Duncavage; Timothy L. Mosbruger; Philippe Szankasi; Todd W. Kelley

OBJECTIVES To design and evaluate a next-generation sequencing (NGS)-based method for T-cell receptor γ (TCRG) gene-based T-cell clonality testing on the Ion Torrent Personal Genome Machine (Life Technologies, Carlsbad, CA) platform. METHODS We analyzed a series of peripheral blood, bone marrow, and formalin-fixed paraffin-embedded tissue specimens with NGS vs traditional capillary electrophoresis methods. RESULTS Using a custom analysis algorithm that we developed, our NGS assay identified between 2,215 and 48,222 unique TCRG rearrangements in a series of 48 samples. We established criteria for assigning clonality based on parameters derived from both the relative and absolute frequencies of reads. In a comparison with standard capillary electrophoresis, 19 of 19 polyclonal samples and 24 of 27 samples that appeared clonal were in agreement. The three discrepant samples demonstrated some of the pitfalls of amplicon length-based testing. Dilution studies with T-lymphoid cell lines demonstrated that a known clonal sequence could be routinely identified when present in as few as 0.1% of total cells demonstrating suitability in residual disease testing. A series of samples was also analyzed on a second NGS platform and yielded very similar results with respect to the frequency and sequence of the clonal rearrangement. CONCLUSIONS In this proof-of-concept study, we describe an NGS-based T-cell clonality assay that is suitable for routine clinical testing either alone or as an adjunct to traditional methods.


Blood | 2017

miR-155 promotes FLT3-ITD–induced myeloproliferative disease through inhibition of the interferon response

Jared Wallace; Dominique A. Kagele; Anna M. Eiring; Carissa N. Kim; Ruozhen Hu; Marah C. Runtsch; Margaret Alexander; Thomas B. Huffaker; Soh Hyun Lee; Ami B. Patel; Timothy L. Mosbruger; Dinesh S. Rao; Rodney R. Miles; June L. Round; Michael W. Deininger; Ryan M. O'Connell

FLT3-ITD+ acute myeloid leukemia (AML) accounts for ∼25% of all AML cases and is a subtype that carries a poor prognosis. microRNA-155 (miR-155) is specifically overexpressed in FLT3-ITD+ AML compared with FLT3 wild-type (FLT3-WT) AML and is critical for the growth of FLT3-ITD+ AML cells in vitro. However, miR-155s role in regulating FLT3-ITD-mediated disease in vivo remains unclear. In this study, we used a genetic mouse model to determine whether miR-155 influences the development of FLT3-ITD-induced myeloproliferative disease. Results indicate that miR-155 promotes FLT3-ITD-induced myeloid expansion in the bone marrow, spleen, and peripheral blood. Mechanistically, miR-155 increases proliferation of the hematopoietic stem and progenitor cell compartments by reducing the growth-inhibitory effects of the interferon (IFN) response, and this involves targeting of Cebpb. Consistent with our observations in mice, primary FLT3-ITD+ AML clinical samples have significantly higher miR-155 levels and a lower IFN response compared with FLT3-WT AML samples. Further, inhibition of miR-155 in FLT3-ITD+ AML cell lines using CRISPR/Cas9, or primary FLT3-ITD+ AML samples using locked nucleic acid antisense inhibitors, results in an elevated IFN response and reduces colony formation. Altogether, our data reveal that miR-155 collaborates with FLT3-ITD to promote myeloid cell expansion in vivo and that this involves a multitarget mechanism that includes repression of IFN signaling.


Oncotarget | 2018

Reversible LSD1 inhibition with HCI-2509 induces the p53 gene expression signature and disrupts the MYCN signature in highrisk neuroblastoma cells

Sumati Gupta; Kelly Doyle; Timothy L. Mosbruger; Andrew Butterfield; Alexis Weston; Allison Ast; Mohan R. Kaadige; Anupam Verma; Sunil Sharma

Lysine-Specific Demethylase 1 (LSD1) over-expression correlates with poorly differentiated neuroblastoma and predicts poor outcome despite multimodal therapy. We have studied the efficacy of reversible and specific LSD1 inhibition with HCI-2509 in neuroblastoma cell lines and particularly the effect of HCI-2509 on the transcriptomic profile in MYCN amplified NGP cells. Cell survival assays show that HCI-2509 is cytotoxic to poorly differentiated neuroblastoma cell lines in low micromole or lower doses. Transcriptional profiling of NGP cells treated with HCI-2509 shows a significant effect on p53, cell cycle, MYCN and hypoxia pathway gene sets. HCI-2509 results in increased histone methyl marks and p53 levels along with cell cycle arrest in the G2/M phase and inhibition of colony formation of NGP cells. Our findings indicate that LSD1 inhibition with HCI-2509 has a multi-target effect in neuroblastoma cell lines, mediated in part via p53. MYCN-amplified neuroblastoma cells have a targeted benefit as HCI-2509 downregulates the MYCN upregulated gene set.


Cancer Research | 2016

Discordant Haplotype Sequencing Identifies Functional Variants at the 2q33 Breast Cancer Risk Locus

Nicola J. Camp; Wei-Yu Lin; Alex Bigelow; George J. Burghel; Timothy L. Mosbruger; Marina Parry; Rosalie Waller; Sushilaben H. Rigas; Pei Yi Tai; Kristofer C. Berrett; Venkatesh Rajamanickam; Rachel Cosby; Ian W. Brock; Brandt Jones; Dan Connley; Robert Sargent; Guoying Wang; Rachel E. Factor; Philip S. Bernard; Lisa A. Cannon-Albright; Stacey Knight; Ryan Abo; Theresa L. Werner; Malcolm Reed; Jason Gertz; Angela Cox

The findings from genome-wide association studies hold enormous potential for novel insight into disease mechanisms. A major challenge in the field is to map these low-risk association signals to their underlying functional sequence variants (FSV). Simple sequence study designs are insufficient, as the vast numbers of statistically comparable variants and a limited knowledge of noncoding regulatory elements complicate prioritization. Furthermore, large sample sizes are typically required for adequate power to identify the initial association signals. One important question is whether similar sample sizes need to be sequenced to identify the FSVs. Here, we present a proof-of-principle example of an extreme discordant design to map FSVs within the 2q33 low-risk breast cancer locus. Our approach employed DNA sequencing of a small number of discordant haplotypes to efficiently identify candidate FSVs. Our results were consistent with those from a 2,000-fold larger, traditional imputation-based fine-mapping study. To prioritize further, we used expression-quantitative trait locus analysis of RNA sequencing from breast tissues, gene regulation annotations from the ENCODE consortium, and functional assays for differential enhancer activities. Notably, we implicate three regulatory variants at 2q33 that target CASP8 (rs3769823, rs3769821 in CASP8, and rs10197246 in ALS2CR12) as functionally relevant. We conclude that nested discordant haplotype sequencing is a promising approach to aid mapping of low-risk association loci. The ability to include more efficient sequencing designs into mapping efforts presents an opportunity for the field to capitalize on the potential of association loci and accelerate translation of association signals to their underlying FSVs. Cancer Res; 76(7); 1916-25. ©2016 AACR.


Immunity | 2018

The Lineage-Defining Transcription Factors SOX2 and NKX2-1 Determine Lung Cancer Cell Fate and Shape the Tumor Immune Microenvironment.

Gurkan Mollaoglu; Alex Jones; Sarah J. Wait; Anandaroop Mukhopadhyay; Sangmin Jeong; Rahul Arya; Soledad A. Camolotto; Timothy L. Mosbruger; Chris J. Stubben; Christopher J. Conley; Arjun Bhutkar; Jeffery M. Vahrenkamp; Kristofer C. Berrett; Melissa Cessna; Thomas E. Lane; Benjamin L. Witt; Mohamed E. Salama; Jason Gertz; Kevin B. Jones; Eric L. Snyder; Trudy G. Oliver

Graphical Abstract Figure. No caption available. SUMMARY The major types of non‐small‐cell lung cancer (NSCLC)—squamous cell carcinoma and adenocarcinoma—have distinct immune microenvironments. We developed a genetic model of squamous NSCLC on the basis of overexpression of the transcription factor Sox2, which specifies lung basal cell fate, and loss of the tumor suppressor Lkb1 (SL mice). SL tumors recapitulated gene‐expression and immune‐infiltrate features of human squamous NSCLC; such features included enrichment of tumor‐associated neutrophils (TANs) and decreased expression of NKX2–1, a transcriptional regulator that specifies alveolar cell fate. In Kras‐driven adenocarcinomas, mis‐expression of Sox2 or loss of Nkx2–1 led to TAN recruitment. TAN recruitment involved SOX2‐mediated production of the chemokine CXCL5. Deletion of Nkx2–1 in SL mice (SNL) revealed that NKX2–1 suppresses SOX2‐driven squamous tumorigenesis by repressing adeno‐to‐squamous transdifferentiation. Depletion of TANs in SNL mice reduced squamous tumors, suggesting that TANs foster squamous cell fate. Thus, lineage‐defining transcription factors determine the tumor immune microenvironment, which in turn might impact the nature of the tumor. HIGHLIGHTSMouse models of lung cancer recapitulate human NSCLC immune microenvironmentSOX2 suppresses NKX2–1 activity, and NKX2–1 represses TAN recruitmentNKX2–1 loss accelerates adeno‐to‐squamous transdifferentiationTANs possess tumor‐promoting features and impact squamous tumorigenesis &NA; Mollaoglu et al. use genetically engineered mouse models of non‐small‐cell lung cancer (NSCLC) to define the relationship between lineage‐defining transcription factors and squamous NSCLC and adenocarcinoma, revealing that this interplay also determines neutrophil recruitment. Depletion of tumor‐associated neutrophils (TANs) reduced squamous tumors and promoted adenocarcinoma features, suggesting that the immune microenvironment might contribute to the nature of the tumor.


bioRxiv | 2017

Nkx2.5-dependent alterations of the embryonic heart DNA methylome identify novel cis-regulatory elements in cardiac development

Bushra Gorsi; Timothy L. Mosbruger; Megan Smith; Jonathon T. Hill; H. Joseph Yost

Transcription factor Nkx2.5 is frequently mutated in congenital heart disease, but the mechanisms by which Nkx2.5 regulates heart development are poorly understood. By generating comprehensive DNA methylome maps from zebrafish embryonic hearts in nxk2.5 mutants and siblings, we discovered that Nkx2.5 regulates DNA methylation patterns during cardiac morphogenesis. We identified hundreds of Nkx-dependent heart-specific Differentially Methylated Regions (nhDMRs). A majority of the nhDMRs were hypomethylated in nkx2.5−/- hearts, correlating with changes in the mutant transcriptome, suggesting Nkx2.5 functions largely as a repressor. Distinct Nkx DNA-binding motifs were significantly enriched in subclasses of nhDMRs. Furthermore, nhDMRs were significantly associated with histone H3K4me1 and H3K27ac post-translational modifications, suggesting Nkx2.5 regulates gene expression by differential methylation of cis-regulatory elements. Using transgenics, we validated several nhDMRs with enhancer activities in the heart. We propose a novel role of Nkx2.5 mediated DNA methylation is integral in activating and repressing Nkx2.5 target genes during heart development.


Journal of the Endocrine Society | 2017

POLR2C Mutations Are Associated With Primary Ovarian Insufficiency in Women

Mika Moriwaki; Barry Moore; Timothy L. Mosbruger; Deborah W. Neklason; Mark Yandell; Lynn B. Jorde; Corrine K. Welt

CONTEXT Primary ovarian insufficiency (POI) results from a premature loss of oocytes, causing infertility and early menopause. The etiology of POI remains unknown in a majority of cases. OBJECTIVE To identify candidate genes in families affected by POI. DESIGN This was a family-based genetic study. SETTING The study was performed at two academic institutions. PATIENTS AND OTHER PARTICIPANTS A family with four generations of women affected by POI (n = 5). Four of these women, three with an associated autoimmune diagnosis, were studied. The controls (n = 387) were recruited for health in old age. INTERVENTION Whole-genome sequencing was performed. MAIN OUTCOME MEASURE Candidate genes were identified by comparing gene mutations in three family members and 387 control subjects analyzed simultaneously using the pedigree Variant Annotation, Analysis and Search Tool. Data were also compared with that in publicly available databases. RESULTS We identified a heterozygous nonsense mutation in a subunit of RNA polymerase II (POLR2C) that synthesizes messenger RNA. A rare sequence variant in POLR2C was also identified in one of 96 women with sporadic POI. POLR2C expression was decreased in the proband compared with women with POI from another cause. Knockdown in an embryonic carcinoma cell line resulted in decreased protein production and impaired cell proliferation. CONCLUSIONS These data support a role for RNA polymerase II mutations as candidates in the etiology of POI. The current data also support results from genome-wide association studies that hypothesize a role for RNA polymerase II subunits in age at menopause in the population.Context: Primary ovarian insufficiency (POI) results from a premature loss of oocytes, causing infertility and early menopause. The etiology of POI remains unknown in a majority of cases. Objective: To identify candidate genes in families affected by POI. Design: This was a family-based genetic study. Setting: The study was performed at two academic institutions. Patients and Other Participants: A family with four generations of women affected by POI (n = 5). Four of these women, three with an associated autoimmune diagnosis, were studied. The controls (n = 387) were recruited for health in old age. Intervention: Whole-genome sequencing was performed. Main Outcome Measure: Candidate genes were identified by comparing gene mutations in three family members and 387 control subjects analyzed simultaneously using the pedigree Variant Annotation, Analysis and Search Tool. Data were also compared with that in publicly available databases. Results: We identified a heterozygous nonsense mutation in a subunit of RNA polymerase II (POLR2C) that synthesizes messenger RNA. A rare sequence variant in POLR2C was also identified in one of 96 women with sporadic POI. POLR2C expression was decreased in the proband compared with women with POI from another cause. Knockdown in an embryonic carcinoma cell line resulted in decreased protein production and impaired cell proliferation. Conclusions: These data support a role for RNA polymerase II mutations as candidates in the etiology of POI. The current data also support results from genome-wide association studies that hypothesize a role for RNA polymerase II subunits in age at menopause in the population.


Cancer Cell | 2017

MYC Drives Progression of Small Cell Lung Cancer to a Variant Neuroendocrine Subtype with Vulnerability to Aurora Kinase Inhibition

Gurkan Mollaoglu; Matthew R. Guthrie; Stefanie Böhm; Johannes Brägelmann; Ismail Can; Paul M. Ballieu; Annika Marx; Julie George; Christine Heinen; Milind D. Chalishazar; Haixia Cheng; Abbie S. Ireland; Kendall E. Denning; Anandaroop Mukhopadhyay; Jeffery M. Vahrenkamp; Kristofer C. Berrett; Timothy L. Mosbruger; Jun Wang; Jessica L. Kohan; Mohamed E. Salama; Benjamin L. Witt; Martin Peifer; Roman K. Thomas; Jason Gertz; Jane E. Johnson; Adi F. Gazdar; Robert J. Wechsler-Reya; Martin L. Sos; Trudy G. Oliver

Collaboration


Dive into the Timothy L. Mosbruger's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dinesh S. Rao

University of California

View shared research outputs
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