Network


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

Hotspot


Dive into the research topics where Jared J. Barrott is active.

Publication


Featured researches published by Jared J. Barrott.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Deletion of mouse Porcn blocks Wnt ligand secretion and reveals an ectodermal etiology of human focal dermal hypoplasia/Goltz syndrome

Jared J. Barrott; Gabriela M. Cash; Aaron P. Smith; Jeffery R. Barrow; L. Charles Murtaugh

The Drosophila porcupine gene is required for secretion of wingless and other Wnt proteins, and sporadic mutations in its unique human ortholog, PORCN, cause a pleiotropic X-linked dominant disorder, focal dermal hypoplasia (FDH, also known as Goltz syndrome). We generated a conditional allele of the X-linked mouse Porcn gene and analyzed its requirement in Wnt signaling and embryonic development. We find that Porcn-deficient cells exhibit a cell-autonomous defect in Wnt ligand secretion but remain responsive to exogenous Wnts. Consistent with the female-specific inheritance pattern of FDH, Porcn hemizygous male embryos arrest during early embryogenesis and fail to generate mesoderm, a phenotype previously associated with loss of Wnt activity. Heterozygous Porcn mutant females exhibit a spectrum of limb, skin, and body patterning abnormalities resembling those observed in human patients with FDH. Many of these defects are recapitulated by ectoderm-specific deletion of Porcn, substantiating a long-standing hypothesis regarding the etiology of human FDH and extending previous studies that have focused on downstream elements of Wnt signaling, such as β-catenin. Conditional deletion of Porcn thus provides an experimental model of FDH, as well as a valuable tool to probe Wnt ligand function in vivo.


Oncogene | 2016

The impact of chromosomal translocation locus and fusion oncogene coding sequence in synovial sarcomagenesis

Kevin B. Jones; Jared J. Barrott; Mingchao Xie; Malay Haldar; Huifeng Jin; Ju Fen Zhu; Michael J. Monument; Tim Mosbruger; Ellen M. Langer; Randall Rl; Richard Wilson; Bradley R. Cairns; Li Ding; Mario R. Capecchi

Synovial sarcomas are aggressive soft-tissue malignancies that express chromosomal translocation-generated fusion genes, SS18-SSX1 or SS18-SSX2 in most cases. Here, we report a mouse sarcoma model expressing SS18-SSX1, complementing our prior model expressing SS18-SSX2. Exome sequencing identified no recurrent secondary mutations in tumors of either genotype. Most of the few mutations identified in single tumors were present in genes that were minimally or not expressed in any of the tumors. Chromosome 6, either entirely or around the fusion gene expression locus, demonstrated a copy number gain in a majority of tumors of both genotypes. Thus, by fusion oncogene coding sequence alone, SS18-SSX1 and SS18-SSX2 can each drive comparable synovial sarcomagenesis, independent from other genetic drivers. SS18-SSX1 and SS18-SSX2 tumor transcriptomes demonstrated very few consistent differences overall. In direct tumorigenesis comparisons, SS18-SSX2 was slightly more sarcomagenic than SS18-SSX1, but equivalent in its generation of biphasic histologic features. Meta-analysis of human synovial sarcoma patient series identified two tumor–gentoype–phenotype correlations that were not modeled by the mice, namely a scarcity of male hosts and biphasic histologic features among SS18-SSX2 tumors. Re-analysis of human SS18-SSX1 and SS18-SSX2 tumor transcriptomes demonstrated very few consistent differences, but highlighted increased native SSX2 expression in SS18-SSX1 tumors. This suggests that the translocated locus may drive genotype–phenotype differences more than the coding sequence of the fusion gene created. Two possible roles for native SSX2 in synovial sarcomagenesis are explored. Thus, even specific partial failures of mouse genetic modeling can be instructive to human tumor biology.


PLOS ONE | 2017

HDAC and proteasome inhibitors synergize to activate pro-apoptotic factors in synovial sarcoma

Aimeâe N. Laporte; Jared J. Barrott; Ren Jie Yao; Neal Poulin; Bertha Brodin; Kevin B. Jones; T. Michael Underhill; Torsten O. Nielsen

Conventional cytotoxic therapies for synovial sarcoma provide limited benefit, and no drugs specifically targeting its driving SS18-SSX fusion oncoprotein are currently available. Patients remain at high risk for early and late metastasis. A high-throughput drug screen consisting of over 900 tool compounds and epigenetic modifiers, representing over 100 drug classes, was undertaken in a panel of synovial sarcoma cell lines to uncover novel sensitizing agents and targetable pathways. Top scoring drug categories were found to be HDAC inhibitors and proteasomal targeting agents. We find that the HDAC inhibitor quisinostat disrupts the SS18-SSX driving protein complex, thereby reestablishing expression of EGR1 and CDKN2A tumor suppressors. In combination with proteasome inhibition, HDAC inhibitors synergize to decrease cell viability and elicit apoptosis. Quisinostat inhibits aggresome formation in response to proteasome inhibition, and combination treatment leads to elevated endoplasmic reticulum stress, activation of pro-apoptotic effector proteins BIM and BIK, phosphorylation of BCL-2, increased levels of reactive oxygen species, and suppression of tumor growth in a murine model of synovial sarcoma. This study identifies and provides mechanistic support for a particular susceptibility of synovial sarcoma to the combination of quisinostat and proteasome inhibition.


Journal of Experimental Medicine | 2016

Modeling synovial sarcoma metastasis in the mouse: PI3′-lipid signaling and inflammation

Jared J. Barrott; Lisa A. Kafchinski; Huifeng Jin; Jared W. Potter; Sarmishta Diraviam Kannan; Robert Kennedy; Tim Mosbruger; Wei Lien Wang; Jen Wei Tsai; Dejka M. Araujo; Ting Liu; Mario R. Capecchi; Alexander J. Lazar; Kevin B. Jones

Barrott et al. show that PI3′-lipid signaling potentiates metastasis in a genetically engineered mouse model of synovial sarcomagenesis and drives cancer cells to express CSF1, recruiting macrophages to the tumor microenvironment.


Molecular Cancer Research | 2017

The Influential Role of BCL2 Family Members in Synovial Sarcomagenesis

Jared J. Barrott; Ju Fen Zhu; Kyllie Smith-Fry; Asia M. Susko; Dakota Nollner; Lance D. Burrell; Amir Pozner; Mario R. Capecchi; Jeffrey T. Yap; Lisa A. Cannon-Albright; Xingming Deng; Kevin B. Jones

Synovial sarcomas are deadly soft tissue malignancies associated with t(X;18) balanced chromosomal translocations. Expression of the apoptotic regulator BCL2 is prominent in synovial sarcomas and has prompted the hypothesis that synovial sarcomagenesis may depend on it. Herein, it is demonstrated that Bcl2 overexpression enhances synovial sarcomagenesis in an animal model. Furthermore, we determined increased familial clustering of human synovial sarcoma patients with victims of other BCL2-associated malignancies in the Utah Population Database. Conditional genetic disruption of Bcl2 in mice also led to reduced sarcomagenesis. Pharmacologic inhibition specific to BCL2 had no demonstrable efficacy against human synovial sarcoma cell lines or mouse tumors. However, targeting BCLxL in human and mouse synovial sarcoma with the small molecule BH3 domain inhibitor, BXI-72, achieved significant cytoreduction and increased apoptotic signaling. Thus, the contributory role of BCL2 in synovial sarcomagenesis does not appear to render it as a therapeutic target, but mitochondrial antiapoptotic BCL2 family members may be. Implications: The association of BCL2 expression with synovial sarcoma is found to fit with a subtle, but significant, impact of its enhanced presence or absence during early tumorigenesis. However, specific pharmacologic inhibition of BCL2 does not demonstrate a persistent dependence in fully developed tumors. Conversely, inhibition of the BCL2 family member BCLxL resulted in nanomolar potency against human synovial sarcoma cell lines and 50% tumor reduction in a genetically engineered mouse model. Mol Cancer Res; 15(12); 1733–40. ©2017 AACR.


Translational Oncology | 2016

LRP5 Signaling in Osteosarcomagenesis: a Cautionary Tale of Translation from Cell Lines to Tumors

Logan Horne; Frank R. Avilucea; Huifeng Jin; Jared J. Barrott; Kyllie Smith-Fry; Yanliang Wang; Bang H. Hoang; Kevin B. Jones

Previous reports document expression of low-density lipoprotein receptor-related protein 5 (LRP5) in osteosarcoma (OS) tissue. Expression of this Wnt receptor correlated with metastatic disease and poor disease-free survival. Forced expression of dominant-negative LRP5 (dnLRP5), which lacks the membrane binding domain of the native protein and therefore functions as a soluble receptor-sponge for Wnt ligands, reduced in vitro cellular invasion and in vivo xenograft tumor growth for osteosarcoma cell lines. Here, we use a genetically engineered mouse model of osteosarcomagenesis with and without expression of dnLRP5 to assess to what degree tumorigenesis is affected and whether Wnt/β-catenin signaling is circumvented or maintained. Each cohort of mice developed osteosarcoma at a similar ultimate prevalence, but after a slightly increased latency in those also expressing dnLRP5. On histology, there was no difference between groups, despite previous reports that the dnLRP5 osteosarcoma cells specifically undergo a mesenchymal-to-epithelial transition in vitro. Finally, immunohistochemistry showed the presence of cytosolic and nuclear β-catenin and nuclear Cyclin D1, markers consistent with preserved Wnt/β-catenin signaling despite constitutive blockade of the cell surface receipt of Wnt signaling ligand. These data suggest that canonical Wnt signaling plays a role in OS progression and that while blockade of singular nodes in signaling pathways can have dramatic effects on individual cell lines, real tumors readily evade such focused attacks.


Molecular Cancer Therapeutics | 2017

Death by HDAC inhibition in synovial sarcoma cells

Aimée N. Laporte; Neal Poulin; Jared J. Barrott; Xiu Qing Wang; Alireza Lorzadeh; Ryan Vander Werff; Kevin B. Jones; T. Michael Underhill; Torsten O. Nielsen

Conventional cytotoxic therapies for synovial sarcoma provide limited benefit, and no drugs specifically targeting the causative SS18-SSX fusion oncoprotein are currently available. Histone deacetylase (HDAC) inhibition has been shown in previous studies to disrupt the synovial sarcoma oncoprotein complex, resulting in apoptosis. To understand the molecular effects of HDAC inhibition, RNA-seq transcriptome analysis was undertaken in six human synovial sarcoma cell lines. HDAC inhibition induced pathways of cell-cycle arrest, neuronal differentiation, and response to oxygen-containing species, effects also observed in other cancers treated with this class of drugs. More specific to synovial sarcoma, polycomb group targets were reactivated, including tumor suppressor CDKN2A, and proapoptotic transcriptional patterns were induced. Functional analyses revealed that ROS-mediated FOXO activation and proapoptotic factors BIK, BIM, and BMF were important to apoptosis induction following HDAC inhibition in synovial sarcoma. HDAC inhibitor pathway activation results in apoptosis and decreased tumor burden following a 7-day quisinostat treatment in the Ptenfl/fl;hSS2 mouse model of synovial sarcoma. This study provides mechanistic support for a particular susceptibility of synovial sarcoma to HDAC inhibition as a means of clinical treatment. Mol Cancer Ther; 16(12); 2656–67. ©2017 AACR.


Journal of Clinical Investigation | 2017

Paracrine osteoprotegerin and β-catenin stabilization support synovial sarcomagenesis in periosteal cells

Jared J. Barrott; Benjamin E. Illum; Huifeng Jin; Matthew L. Hedberg; Yanliang Wang; Allie H. Grossmann; Malay Haldar; Mario R. Capecchi; Kevin B. Jones

Synovial sarcoma (SS) is an aggressive soft-tissue sarcoma that is often discovered during adolescence and young adulthood. Despite the name, synovial sarcoma does not typically arise from a synoviocyte but instead arises in close proximity to bones. Previous work demonstrated that mice expressing the characteristic SS18-SSX fusion oncogene in myogenic factor 5–expressing (Myf5-expressing) cells develop fully penetrant sarcomagenesis, suggesting skeletal muscle progenitor cell origin. However, Myf5 is not restricted to committed myoblasts in embryos but is also expressed in multipotent mesenchymal progenitors. Here, we demonstrated that human SS and mouse tumors arising from SS18-SSX expression in the embryonic, but not postnatal, Myf5 lineage share an anatomic location that is frequently adjacent to bone. Additionally, we showed that SS can originate from periosteal cells expressing SS18-SSX alone and from preosteoblasts expressing the fusion oncogene accompanied by the added stabilization of &bgr;-catenin, which is a common secondary change in SS. Expression and secretion of the osteoclastogenesis inhibitory factor osteoprotegerin enabled early growth of SS18-SSX2–transformed cells, indicating a paracrine link between the bone and synovial sarcomagenesis. These findings explain the skeletal contact frequently observed in human SS and may provide alternate means of enabling SS18-SSX–driven oncogenesis in cells as differentiated as preosteoblasts.


Cancer Microenvironment | 2017

The Impact of Microenvironment on the Synovial Sarcoma Transcriptome

Huifeng Jin; Jared J. Barrott; Matthew G. Cable; Michael J. Monument; Daniel M. Lerman; Kyllie Smith-Fry; Dakota Nollner; Kevin B. Jones

Synovial sarcoma (SS) is initiated by a t(X;18) chromosomal translocation and resultant SS18-SSX fusion oncogene. Only a few SS cell lines exist. None has been compared to its source tumor. In order to compare matched tumor and cell line pairs, we performed RNAseq on 3 tumor/cell line pairs from a genetically engineered mouse model of SS, as well as 2 pairs from human SS tumors. Transcriptomes of mouse tumors and derivative cell lines deviated significantly. Differentially expressed genes highlighted inflammatory infiltrates and metabolism. The same was found for the human tumor and cell line pairs. More was shared between different tumors than between any tumor and its cell line. Direct xenografting generated transcriptomes that more closely resembled the primary tumor than did its derivative cell line. SS tumor transcriptomes are powerfully impacted by the environment wherein they reside, especially with regard to immune interaction and metabolism.


Clinical Cancer Research | 2016

Abstract 03: The impact of chromosomal translocation locus and fusion oncogene coding sequence in synovial sarcomagenesis

Kevin B. Jones; Jared J. Barrott; Huifeng Jin; Malay Haldar; Mario R. Capecchi

Synovial sarcoma, the most common soft-tissue sarcoma in young adults bears a t(X;18) translocation that generates a fusion between SS18 (formerly SYT) and an SSX gene. SS18-SSX1 and SS18-SSX2 differ by only 13 amino acids. Clinically, SS18-SSX1 is more common, portends a worse prognosis, and demonstrates increased prevalence of biphasic histology. We conditionally expressed each fusion in otherwise identical mouse models, unexpectedly finding SS18-SSX2 to be more sarcomagenic than SS18-SSX1 and equally prone to biphasic histology. This suggested that coding sequence does not account for clinical differences between the fusions. Reanalysis of patient series found a gender bias in the prevalence of SS18-SSX2-expressing tumors, suggesting an X-linked effect and implicating the loss of native SSX2 by translocation as a hinderance to SS18-SSX2-driven synovial sarcomagenesis in males. Next, we confirmed that human synovial sarcomas almost universally express native SSX2, but not SSX1 and knock-down of native SSX2 in human synovial sarcoma cell lines blunted proliferation whereas overexpression of SSX2 enhances cell proliferation. In conclusion, we hypothesize that SSX2 facilitates sarcomagenesis in SS18-SSX1-expressing tumors and because our model of SS18-SSX2-expressing mice does not disrupt native SSX2 it recapitulates the prevalence and phenotype of SS18-SSX1 tumors. Citation Format: Kevin B. Jones, Jared J. Barrott, Huifeng Jin, Malay Haldar, Mario R. Capecchi. The impact of chromosomal translocation locus and fusion oncogene coding sequence in synovial sarcomagenesis. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Integrating Clinical Genomics and Cancer Therapy; Jun 13-16, 2015; Salt Lake City, UT. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(1_Suppl):Abstract nr 03.

Collaboration


Dive into the Jared J. Barrott'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

Malay Haldar

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neal Poulin

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge