Network


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

Hotspot


Dive into the research topics where Jacob A. Herman is active.

Publication


Featured researches published by Jacob A. Herman.


Nucleic Acids Research | 2013

Linker histone H1.0 interacts with an extensive network of proteins found in the nucleolus

Anna A. Kalashnikova; Duane D. Winkler; Steven J. McBryant; Ryan K. Henderson; Jacob A. Herman; Jennifer G. DeLuca; Karolin Luger; Jessica E. Prenni; Jeffrey C. Hansen

The H1 linker histones are abundant chromatin-associated DNA-binding proteins. Recent evidence suggests that linker histones also may function through protein–protein interactions. To gain a better understanding of the scope of linker histone involvement in protein–protein interactions, we used a proteomics approach to identify H1-binding proteins in human nuclear extracts. Full-length H1.0 and H1.0 lacking its C-terminal domain (CTD) were used for protein pull-downs. A total of 107 candidate H1.0 binding proteins were identified by LC-MS/MS. About one-third of the H1.0-dependent interactions were mediated by the CTD, and two-thirds by the N-terminal domain-globular domain fragment. Many of the proteins pulled down by H1.0 were core splicing factors. Another group of H1-binding proteins functions in rRNA biogenesis. H1.0 also pulled down numerous ribosomal proteins and proteins involved in cellular transport. Strikingly, nearly all of the H1.0-binding proteins are found in the nucleolus. Quantitative biophysical studies with recombinant proteins confirmed that H1.0 directly binds to FACT and the splicing factors SF2/ASF and U2AF65. Our results demonstrate that H1.0 interacts with an extensive network of proteins that function in RNA metabolism in the nucleolus, and suggest that a new paradigm for linker histone action is in order.


Cancer Discovery | 2013

Cancer-Specific Requirement for BUB1B/BUBR1 in Human Brain Tumor Isolates and Genetically Transformed Cells

Yu Ding; Christopher G. Hubert; Jacob A. Herman; Philip Corrin; Chad M. Toledo; Kyobi Skutt-Kakaria; Julio Vazquez; Ryan Basom; Bin Zhang; Jennifer K. Risler; Steven M. Pollard; Do Hyun Nam; Jeffery J. Delrow; Jun Zhu; Jeongwu Lee; Jennifer G. DeLuca; James M. Olson; Patrick J. Paddison

UNLABELLED To identify new candidate therapeutic targets for glioblastoma multiforme, we combined functional genetics and glioblastoma network modeling to identify kinases required for the growth of patient-derived brain tumor-initiating cells (BTIC) but that are dispensable to proliferating human neural stem cells (NSC). This approach yielded BUB1B/BUBR1, a critical mitotic spindle checkpoint player, as the top-scoring glioblastoma lethal kinase. Knockdown of BUB1B inhibited expansion of BTIC isolates, both in vitro and in vivo, without affecting proliferation of NSCs or astrocytes. Mechanistic studies revealed that BUB1Bs GLE2p-binding sequence (GLEBS) domain activity is required to suppress lethal kinetochore-microtubule (KT-MT) attachment defects in glioblastoma isolates and genetically transformed cells with altered sister KT dynamics, which likely favor KT-MT instability. These results indicate that glioblastoma tumors have an added requirement for BUB1B to suppress lethal consequences of altered KT function and further suggest that sister KT measurements may predict cancer-specific sensitivity to BUB1B inhibition and perhaps other mitotic targets that affect KT-MT stability. SIGNIFICANCE Currently, no effective therapies are available for glioblastoma, the most frequent and aggressive brain tumor. Our results suggest that targeting the GLEBS domain activity of BUB1B may provide a therapeutic window for glioblastoma, as the GLEBS domain is nonessential in untransformed cells. Moreover, the results further suggest that sister KT distances at metaphase may predict sensitivity to anticancer therapeutics targeting KT function.


Journal of Molecular Biology | 2015

Proteomic characterization of the nucleolar linker histone H1 interaction network.

Heather J. Szerlong; Jacob A. Herman; Christine Krause; Jennifer G. DeLuca; Arthur I. Skoultchi; Quinton A. Winger; Jessica E. Prenni; Jeffrey C. Hansen

To investigate the relationship between linker histone H1 and protein-protein interactions in the nucleolus, we used biochemical and proteomics approaches to characterize nucleoli purified from cultured human and mouse cells. Mass spectrometry identified 175 proteins in human T cell nucleolar extracts that bound to Sepharose-immobilized H1 in vitro. Gene ontology analysis found significant enrichment for H1 binding proteins with functions related to nucleolar chromatin structure and RNA polymerase I transcription regulation, rRNA processing, and mRNA splicing. Consistent with the affinity binding results, H1 existed in large (400 to >650kDa) macromolecular complexes in human T cell nucleolar extracts. To complement the biochemical experiments, we investigated the effects of in vivo H1 depletion on protein content and structural integrity of the nucleolus using the H1 triple isoform knockout (H1ΔTKO) mouse embryonic stem cell (mESC) model system. Proteomic profiling of purified wild-type mESC nucleoli identified a total of 613 proteins, only ~60% of which were detected in the H1 mutant nucleoli. Within the affected group, spectral counting analysis quantitated 135 specific nucleolar proteins whose levels were significantly altered in H1ΔTKO mESC. Importantly, the functions of the affected proteins in mESC closely overlapped with those of the human T cell nucleolar H1 binding proteins. Immunofluorescence microscopy of intact H1ΔTKO mESC demonstrated both a loss of nucleolar RNA content and altered nucleolar morphology resulting from in vivo H1 depletion. We conclude that H1 organizes and maintains an extensive protein-protein interaction network in the nucleolus required for nucleolar structure and integrity.


Clinical Cancer Research | 2015

Molecular Pathways: Regulation and Targeting of Kinetochore–Microtubule Attachment in Cancer

Jacob A. Herman; Chad M. Toledo; James M. Olson; Jennifer G. DeLuca; Patrick J. Paddison

Kinetochores are large protein structures assembled on centromeric DNA during mitosis that bind to microtubules of the mitotic spindle to orchestrate and power chromosome movements. Deregulation of kinetochore–microtubule (KT–MT) attachments has been implicated in driving chromosome instability and cancer evolution; however, the nature and source of KT–MT attachment defects in cancer cells remain largely unknown. Here, we highlight recent findings suggesting that oncogene-driven changes in kinetochore regulation occur in glioblastoma multiforme (GBM) and possibly other cancers exhibiting chromosome instability, giving rise to novel therapeutic opportunities. In particular, we consider the GLE2p-binding sequence domains of BubR1 and the newly discovered BuGZ, two kinetochore-associated proteins, as candidate therapeutic targets for GBM. Clin Cancer Res; 21(2); 233–9. ©2014 AACR.


Cancer Research | 2017

Sensitivity to BUB1B Inhibition Defines an Alternative Classification of Glioblastoma

Eunjee Lee; Margaret Pain; Huaien Wang; Jacob A. Herman; Chad M. Toledo; Jennifer G. DeLuca; Raymund Yong; Patrick J. Paddison; Jun Zhu

Glioblastoma multiforme (GBM) remains a mainly incurable disease in desperate need of more effective treatments. In this study, we develop evidence that the mitotic spindle checkpoint molecule BUB1B may offer a predictive marker for aggressiveness and effective drug response. A subset of GBM tumor isolates requires BUB1B to suppress lethal kinetochore-microtubule attachment defects. Using gene expression data from GBM stem-like cells, astrocytes, and neural progenitor cells that are sensitive or resistant to BUB1B inhibition, we created a computational framework to predict sensitivity to BUB1B inhibition. Applying this framework to tumor expression data from patients, we stratified tumors into BUB1B-sensitive (BUB1BS) or BUB1B-resistant (BUB1BR) subtypes. Through this effort, we found that BUB1BS patients have a significantly worse prognosis regardless of tumor development subtype (i.e., classical, mesenchymal, neural, proneural). Functional genomic profiling of BUB1BR versus BUB1BS isolates revealed a differential reliance of genes enriched in the BUB1BS classifier, including those involved in mitotic cell cycle, microtubule organization, and chromosome segregation. By comparing drug sensitivity profiles, we predicted BUB1BS cells to be more sensitive to type I and II topoisomerase inhibitors, Raf inhibitors, and other drugs, and experimentally validated some of these predictions. Taken together, the results show that our BUB1BR/S classification of GBM tumors can predict clinical course and sensitivity to drug treatment. Cancer Res; 77(20); 5518-29. ©2017 AACR.


Oncotarget | 2017

ZNF131 suppresses centrosome fragmentation in glioblastoma stem-like cells through regulation of HAUS5

Yu Ding; Jacob A. Herman; Chad M. Toledo; Jackie M. Lang; Philip Corrin; Emily J. Girard; Ryan Basom; Jeffrey J. Delrow; James M. Olson; Patrick J. Paddison

Zinc finger domain genes comprise ∼3% of the human genome, yet many of their functions remain unknown. Here we investigated roles for the vertebrate-specific BTB domain zinc finger gene ZNF131 in the context of human brain tumors. We report that ZNF131 is broadly required for Glioblastoma stem-like cell (GSC) viability, but dispensable for neural progenitor cell (NPC) viability. Examination of gene expression changes after ZNF131 knockdown (kd) revealed that ZNF131 activity notably promotes expression of Joubert Syndrome ciliopathy genes, including KIF7, NPHP1, and TMEM237, as well as HAUS5, a component of Augmin/HAUS complex that facilitates microtubule nucleation along the mitotic spindle. Of these genes only kd of HAUS5 displayed GSC-specific viability loss. Critically, HAUS5 ectopic expression was sufficient to suppress viability defects of ZNF131 kd cells. Moreover, ZNF131 and HAUS5 kd phenocopied each other in GSCs, each causing: mitotic arrest, centrosome fragmentation, loss of Augmin/HAUS complex on the mitotic spindle, and loss of GSC self-renewal and tumor formation capacity. In control NPCs, we observed centrosome fragmentation and lethality only when HAUS5 kd was combined with kd of HAUS2 or HAUS4, demonstrating that the complex is essential in NPCs, but that GSCs have heightened requirement. Our results suggest that GSCs differentially rely on ZNF131-dependent expression of HAUS5 as well as the Augmin/HAUS complex activity to maintain the integrity of centrosome function and viability.


Methods of Molecular Biology | 2016

Measuring Kinetochore-Microtubule Attachment Stability in Cultured Cells.

Keith F. DeLuca; Jacob A. Herman; Jennifer G. DeLuca

Duplicated sister chromatids connect to the mitotic spindle through kinetochores, large proteinaceous structures built at sites of centromeric heterochromatin. Kinetochores are responsible for harnessing the forces generated by microtubule polymerization and depolymerization to power chromosome movements. The fidelity of chromosome segregation relies on proper kinetochore function, as precise regulation of the attachment between kinetochores and microtubules is essential to prevent mitotic errors, which are linked to the initiation and progression of cancer and the formation of birth defects (Godek et al., Nat Rev Mol Cell Biol 16(1):57-64, 2014; Ricke and van Deursen, Semin Cell Dev Biol 22(6):559-565, 2011; Holland and Cleveland, EMBO Rep 13(6):501-514, 2012). Here we describe assays to quantitatively measure kinetochore-microtubule attachment stability in cultured cells.


Molecular Biology of the Cell | 2017

Spindle assembly checkpoint signaling and sister chromatid cohesion are disrupted by HPV E6-mediated transformation

Erin P. Kelley; Jennifer G. DeLuca; Jacob A. Herman

HPV-transforming protein E6 weakens both SAC activity and sister chromatid cohesion. These processes largely function normally and result in chromosome segregation errors only when mitosis is delayed by the presence of one to four unaligned, pole-associated chromosomes. This mechanism of aneuploidy is not observed in response to oncogenic signaling.


Molecular Cancer Therapeutics | 2013

Abstract A05: Functional genetic approach identifies cancer-specific requirement for BUB1B/BubR1 in human brain tumor isolates and genetically transformed cells

Yu Ding; Christopher G. Hubert; Jacob A. Herman; Jennifer G. DeLuca; James M. Olson; Patrick J. Paddison

Cancer cells give rise to distinctive and complimentary capabilities that enable them to continuously grow. These capabilities may as well generate additional vulnerabilities and can be targeted for therapy. To identify new candidate therapeutic targets for glioblastoma multiforme, we combined functional genetics and glioblastoma network modeling to identify kinases required for the growth of patient-derived brain tumor-initiating cells (BTIC) but that are dispensable to proliferating human neural stem cells (NSC). This approach yielded BUB1B/BUBR1, a critical mitotic spindle checkpoint player, as the top-scoring glioblastoma lethal kinase. Knockdown of BUB1B inhibited expansion of BTIC isolates, both in vitro and in vivo, without affecting proliferation of NSCs or astrocytes. Mechanistic studies revealed that BUB1B9s GLE2p-binding sequence (GLEBS) domain activity is required to suppress lethal kinetochore-microtubule (KT-MT) attachment defects in glioblastoma isolates and genetically transformed cells with altered sister KT dynamics, which likely favor KT-MT instability. These results indicate that glioblastoma tumors have an added requirement for BUB1B to suppress lethal consequences of altered KT function and further suggest that sister KT measurements may predict cancer-specific sensitivity to BUB1B inhibition and perhaps other mitotic targets that affect KT-MT stability. Significance: Currently, no effective therapies are available for glioblastoma, the most frequent and aggressive brain tumor. Our results suggest that targeting the GLEBS domain activity of BUB1B may provide a therapeutic window for glioblastoma, as the GLEBS domain is nonessential in untransformed cells. Moreover, the results further suggest that sister KT distances at metaphase may predict sensitivity to anticancer therapeutics targeting KT function. Citation Format: Yu Ding, Christopher Hubert, Jacob Herman, Jennifer DeLuca, James Olson, Patrick Paddison. Functional genetic approach identifies cancer-specific requirement for BUB1B/BubR1 in human brain tumor isolates and genetically transformed cells. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Synthetic Lethal Approaches to Cancer Vulnerabilities; May 17-20, 2013; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(5 Suppl):Abstract nr A05.


Genes & Development | 2013

Genome-wide RNAi screens in human brain tumor isolates reveal a novel viability requirement for PHF5A

Christopher G. Hubert; Robert K. Bradley; Yu Ding; Chad M. Toledo; Jacob A. Herman; Kyobi Skutt-Kakaria; Emily J. Girard; Jerry Davison; Jason D. Berndt; Philip Corrin; Justin Hardcastle; Ryan Basom; Jeffery J. Delrow; Thomas R. Webb; Steven M. Pollard; Jeongwu Lee; James M. Olson; Patrick J. Paddison

Collaboration


Dive into the Jacob A. Herman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick J. Paddison

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Chad M. Toledo

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

James M. Olson

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Yu Ding

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Philip Corrin

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Christopher G. Hubert

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Emily J. Girard

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Ryan Basom

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jeffery J. Delrow

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge