Try a new search

Format these results:

Searched for:

person:neelb01

in-biosketch:yes

Total Results:

323


Mechanism for the learning disability in RAS-ERK signaling-associated neurodevelopmental disorder [Meeting Abstract]

Kang, M; Neel, B G; Ryu, H -H; Kim, S; Lee, Y -S
RAS-ERK signaling pathway regulates large numbers of biological processes. Thus, germline mutations in RAS-ERK signaling pathway are associated with developmental disorders, collectively called RASopathy. BRAF is a direct downstream effector of RAS and germline mutations of BRAF accounts for over 70% of cardio-facio-cutaneous (CFC) syndrome which severely impairs cognitive functions. However, the cellular and molecular basis for the brain abnormalities in CFC syndrome remains largely unknown. Here, we investigated how the dysregulated RAS-ERK signaling affects malformation of central nervous system (CNS) and cognitive deficits by using conditional knockin mice harboring RASopathy-associated mutations in Braf. Neural stem cell-specific expression of Braf mutations significantly impaired hippocampus-dependent learning and memory in Morris water maze. Histological examination and transcriptome analyses revealed changes in specific cell types although mutants were expressed in neural stem cells, suggesting that the impacts of expressing Braf mutations are sensitive to specific cell types as previously shown in other RASopathy mouse models. Our study will contribute to untangle the complexity of RAS-ERK signaling network in brain development and cognitive functions.
Copyright
EMBASE:2002731708
ISSN: 2451-8301
CID: 4120422

Activated Thiol Sepharose-based proteomic approach to quantify reversible protein oxidation

Xu, Yang; Andrade, Joshua; Ueberheide, Beatrix; Neel, Benjamin G
Reactive oxygen species (ROS) can act as second messengers in various signaling pathways, and abnormal oxidation contributes to multiple diseases, including cancer. Detecting and quantifying protein oxidation is crucial for a detailed understanding of reduction-oxidation reaction (redox) signaling. We developed an Activated Thiol Sepharose-based proteomic (ATSP) approach to quantify reversible protein oxidation. ATSP can enrich H2O2-sensitive thiol peptides, which are more likely to contain reactive cysteines involved in redox signaling. We applied our approach to analyze hereditary leiomyomatosis and renal cell carcinoma (HLRCC), a type of kidney cancer that harbors fumarate hydratase (FH)-inactivating mutations and has elevated ROS levels. Multiple proteins were oxidized in FH-deficient cells, including many metabolic proteins such as the pyruvate kinase M2 isoform (PKM2). Treatment of HLRCC cells with dimethyl fumarate or PKM2 activators altered PKM2 oxidation levels. Finally, we found that ATSP could detect Src homology region 2 domain-containing phosphatase-2 and PKM2 oxidation in cells stimulated with platelet-derived growth factor. This newly developed redox proteomics workflow can detect reversible oxidation of reactive cysteines and can be employed to analyze multiple physiologic and pathologic conditions.-Xu, Y., Andrade, J., Ueberheide, B., Neel, B. G. Activated Thiol Sepharose-based proteomic approach to quantify reversible protein oxidation.
PMID: 31451050
ISSN: 1530-6860
CID: 4054242

Migration rather than proliferation transcriptomic signatures are strongly associated with breast cancer patient survival

Nair, Nishanth Ulhas; Das, Avinash; Rogkoti, Vasiliki-Maria; Fokkelman, Michiel; Marcotte, Richard; de Jong, Chiaro G; Koedoot, Esmee; Lee, Joo Sang; Meilijson, Isaac; Hannenhalli, Sridhar; Neel, Benjamin G; de Water, Bob van; Le Dévédec, Sylvia E; Ruppin, Eytan
The efficacy of prospective cancer treatments is routinely estimated by in vitro cell-line proliferation screens. However, it is unclear whether tumor aggressiveness and patient survival are influenced more by the proliferative or the migratory properties of cancer cells. To address this question, we experimentally measured proliferation and migration phenotypes across more than 40 breast cancer cell-lines. Based on the latter, we built and validated individual predictors of breast cancer proliferation and migration levels from the cells' transcriptomics. We then apply these predictors to estimate the proliferation and migration levels of more than 1000 TCGA breast cancer tumors. Reassuringly, both estimates increase with tumor's aggressiveness, as qualified by its stage, grade, and subtype. However, predicted tumor migration levels are significantly more strongly associated with patient survival than the proliferation levels. We confirmed these findings by conducting siRNA knock-down experiments on the highly migratory MDA-MB-231 cell lines and deriving gene knock-down based proliferation and migration signatures. We show that cytoskeletal drugs might be more beneficial in patients with high predicted migration levels. Taken together, these results testify to the importance of migration levels in determining patient survival.
PMCID:6662662
PMID: 31358840
ISSN: 2045-2322
CID: 4010732

Genetic targeting of ERK1/ERK2 kinases impairs the fitness of the myeloproliferative neoplasm clone [Meeting Abstract]

Stivala, S; Brkic, S; Dirnhofer, S; Passweg, J R; Tsakiris, D A; Neel, B G; Levine, R L; Meyer, S C
Background: Myeloproliferative neoplasms (MPN) are hematopoietic stem cell disorders with dysregulated JAK2 signaling. The limited effects of JAK inhibitors relate to compensatory MAPK activation. We hypothesized that ERK1/2 could be a favorable target given the distal position in the MAPK pathway and the essential role for hematopoiesis.
Method(s): We genetically targeted ERK1/2 in MPN by combining Jak2V617F with ERK1 and ERK2 knockout alleles and hematopoiesisspecific Mx-Cre.To assess engraftment dynamics and competitive performance of the MPN clone, CD45.2 Jak2V617F bone marrow (BM) +/-ERK1/2 double KO (dKO) was competitively transplanted 1:1 with CD45.1 Jak2 WT BM into WT recipients.
Result(s): Loss of ERK1/2 in Jak2V617F mice reduced splenomegaly at 3 months. Excessive erythropoiesis was moderated with decreased red cell and reticulocyte counts in peripheral blood and erythroid progenitors in BM. Red cell parameters remained slightly above normal over time without induction of anemia. The microcytic hypochromic red cell features of Jak2V617F mice normalized by ERK1/2 dKO and emergence of leukocytosis and neutrophilia was prevented. Platelets were normal in Jak2V617F mice, while ERK1/2 ablation moderated thrombopoiesis with reduced megakaryocyte progenitors and thrombocytopenia. Lin-Sca+Kit+ (LSK) hematopoietic stem/progenitor cells were decreased in ERK deficient Jak2V617F mice suggesting a reduced disease-initiating population. In competitive transplants, all recipients of Jak2V617F ERK1/2 dKO BM engrafted. The Jak2V617F clone as shown by CD45.2 chimerism, was reduced to 19% in mice with deficient vs. intact ERK1/2. The Jak2V617F clone was most prominent in neutrophils and gradually expanded, but remained low and stable in ERK deficient mice.(Figure presented)
Conclusion(s): Our data show that targeting ERK1/2 impairs the fitness of the MPN clone by restricting the stem/progenitor compartment and blunting clone expansion with reduced differentiated cell output and moderated cytoses. While targeting ERK could be a promising combination strategy with JAK2 inhibition, it should be restricted to settings of thrombocytosis to prevent induction of thrombocytopenia
EMBASE:628985197
ISSN: 1424-3997
CID: 4053332

An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity

Kopper, Oded; de Witte, Chris J; Lõhmussaar, Kadi; Valle-Inclan, Jose Espejo; Hami, Nizar; Kester, Lennart; Balgobind, Anjali Vanita; Korving, Jeroen; Proost, Natalie; Begthel, Harry; van Wijk, Lise M; Revilla, Sonia Aristín; Theeuwsen, Rebecca; van de Ven, Marieke; van Roosmalen, Markus J; Ponsioen, Bas; Ho, Victor W H; Neel, Benjamin G; Bosse, Tjalling; Gaarenstroom, Katja N; Vrieling, Harry; Vreeswijk, Maaike P G; van Diest, Paul J; Witteveen, Petronella O; Jonges, Trudy; Bos, Johannes L; van Oudenaarden, Alexander; Zweemer, Ronald P; Snippert, Hugo J G; Kloosterman, Wigard P; Clevers, Hans
Ovarian cancer (OC) is a heterogeneous disease usually diagnosed at a late stage. Experimental in vitro models that faithfully capture the hallmarks and tumor heterogeneity of OC are limited and hard to establish. We present a protocol that enables efficient derivation and long-term expansion of OC organoids. Utilizing this protocol, we have established 56 organoid lines from 32 patients, representing all main subtypes of OC. OC organoids recapitulate histological and genomic features of the pertinent lesion from which they were derived, illustrating intra- and interpatient heterogeneity, and can be genetically modified. We show that OC organoids can be used for drug-screening assays and capture different tumor subtype responses to the gold standard platinum-based chemotherapy, including acquisition of chemoresistance in recurrent disease. Finally, OC organoids can be xenografted, enabling in vivo drug-sensitivity assays. Taken together, this demonstrates their potential application for research and personalized medicine.
PMID: 31011202
ISSN: 1546-170x
CID: 3821462

Phase I study of local radiation and tremelimumab in patients with inoperable locally recurrent or metastatic breast cancer

Jiang, Di Maria; Fyles, Anthony; Nguyen, Linh T; Neel, Benjamin G; Sacher, Adrian; Rottapel, Robert; Wang, Ben X; Ohashi, Pamela S; Sridhar, Srikala S
Immunotherapy has shown modest activity in metastatic breast cancer (MBC). In this phase I dose escalation study, we assessed safety of tremelimumab, a humanized anti-CTLA4 monoclonal antibody, at starting dose 3 mg/kg, on the third day of palliative radiotherapy (2000cGy in 5 daily fractions) in patients with MBC. Primary objective was to determine the maximum tolerated dose (MTD) of tremelimumab combined with RT. Secondary objective was to assess response. Among 6 patients enrolled between July 2010 and October 2011, 5 had hormone receptor-positive MBC, 1 had triple negative MBC. Median age was 45 years. Common toxicities included lymphopenia (83%), fatigue (50%) and rash (33%). One dose-limiting toxicity occurred at 6 mg/kg, however the trial closed before MTD could be determined. One patient discontinued treatment due to a pathological fracture. Best response was stable disease (SD), 1 patient had SD for >6 months. Median follow up was 27.0 months. Median OS was 50.8 months, with 1 patient surviving >8 years. Peripheral blood mononuclear cell (PBMC) profiles showed increasing proliferating (Ki67+) Treg cells 1 week post treatment in 5 patients. Overall, tremelimumab at 3 mg/kg combined with RT appears to be a tolerable treatment strategy. Further studies are needed to optimize this combination approach.
PMCID:6508206
PMID: 31105877
ISSN: 1949-2553
CID: 3898842

The Noonan Syndrome-linked Raf1L613V mutation drives increased glial number in the mouse cortex and enhanced learning

Holter, Michael C; Hewitt, Lauren T; Koebele, Stephanie V; Judd, Jessica M; Xing, Lei; Bimonte-Nelson, Heather A; Conrad, Cheryl D; Araki, Toshiyuki; Neel, Benjamin G; Snider, William D; Newbern, Jason M
RASopathies are a family of related syndromes caused by mutations in regulators of the RAS/Extracellular Regulated Kinase 1/2 (ERK1/2) signaling cascade that often result in neurological deficits. RASopathy mutations in upstream regulatory components, such as NF1, PTPN11/SHP2, and RAS have been well-characterized, but mutation-specific differences in the pathogenesis of nervous system abnormalities remain poorly understood, especially those involving mutations downstream of RAS. Here, we assessed cellular and behavioral phenotypes in mice expressing a Raf1L613V gain-of-function mutation associated with the RASopathy, Noonan Syndrome. We report that Raf1L613V/wt mutants do not exhibit a significantly altered number of excitatory or inhibitory neurons in the cortex. However, we observed a significant increase in the number of specific glial subtypes in the forebrain. The density of GFAP+ astrocytes was significantly increased in the adult Raf1L613V/wt cortex and hippocampus relative to controls. OLIG2+ oligodendrocyte progenitor cells were also increased in number in mutant cortices, but we detected no significant change in myelination. Behavioral analyses revealed no significant changes in voluntary locomotor activity, anxiety-like behavior, or sociability. Surprisingly, Raf1L613V/wt mice mutants performed better than controls in select aspects of the water radial-arm maze, Morris water maze, and cued fear conditioning tasks. Overall, these data show that increased astrocyte and oligodendrocyte progenitor cell (OPC) density in the cortex coincides with enhanced cognition in Raf1L613V/wt mutants and further highlight the distinct effects of RASopathy mutations on nervous system development and function.
PMID: 31017896
ISSN: 1553-7404
CID: 3821652

N-Glycoproteomics of Patient-Derived Xenografts: A Strategy to Discover Tumor-Associated Proteins in High-Grade Serous Ovarian Cancer

Sinha, Ankit; Hussain, Ali; Ignatchenko, Vladimir; Ignatchenko, Alexandr; Tang, Kwan Ho; Ho, Victor W H; Neel, Benjamin G; Clarke, Blaise; Bernardini, Marcus Q; Ailles, Laurie; Kislinger, Thomas
High-grade serous ovarian carcinoma (HGSC) is the most common and lethal subtype of gynecologic malignancy in women. The current standard of treatment combines cytoreductive surgery and chemotherapy. Despite the efficacy of initial treatment, most patients develop cancer recurrence, and 70% of patients die within 5 years of initial diagnosis. CA125 is the current FDA-approved biomarker used in the clinic to monitor response to treatment and recurrence, but its impact on patient survival is limited. New strategies for the discovery of HGSC biomarkers are urgently needed. Here, we describe a proteomics strategy to detect tumor-associated proteins in serum of HGSC patient-derived xenograft models. We demonstrate proof-of-concept applicability using two independent, longitudinal serum cohorts from HGSC patients.
PMID: 30981729
ISSN: 2405-4720
CID: 3810272

The Gp1ba-Cre transgenic mouse: a new model to delineate platelet and leukocyte functions

Nagy, Zoltan; Vögtle, Timo; Geer, Mitchell J; Mori, Jun; Heising, Silke; Di Nunzio, Giada; Gareus, Ralph; Tarakhovsky, Alexander; Weiss, Arthur; Neel, Benjamin G; Desanti, Guillaume E; Mazharian, Alexandra; Senis, Yotis A
Conditional knockout (KO) mouse models are invaluable for elucidating the physiological roles of platelets. The Pf4-Cre transgenic mouse is the current model of choice for generating megakaryocyte/platelet-specific KO mice. Platelets and leukocytes work closely together in a wide range of disease settings, yet the specific contribution of platelets to these processes remains unclear. This is partially due to the Pf4-Cre transgene being expressed in a variety of leukocyte populations. To overcome this issue, we developed a Gp1ba-Cre transgenic mouse strain in which Cre expression in driven by the endogenous Gp1ba locus. By crossing Gp1ba-Cre and Pf4-Cre mice to the mT/mG dual-fluorescence reporter mouse and performing a head-to-head comparison, we demonstrate more stringent megakaryocyte lineage-specific expression of the Gp1ba-Cre transgene. Broader tissue expression was observed with the Pf4-Cre transgene, leading to recombination in many hematopoietic lineages, including monocytes, macrophages, granulocytes, dendritic, B and T cells. Direct comparison of phenotypes of Csk, Shp1 or CD148 conditional KO mice generated using either the Gp1ba-Cre or Pf4-Cre strains revealed similar platelet phenotypes. However, additional inflammatory and immunological anomalies were observed in Pf4-Cre-generated KO mice due to non-specific deletion in other hematopoietic lineages. By excluding leukocyte contributions to phenotypes, the Gp1ba-Cre mouse will advance our understanding of the role of platelets in inflammation and other pathophysiological processes where platelet-leukocyte interactions are involved.
PMID: 30429161
ISSN: 1528-0020
CID: 3457442

The genomic architecture of serous carcinomas shapes the tumor microenvironment and modulates responses to targeted and immunotherapies [Meeting Abstract]

Iyer, S; Zhang, S; Farkkila, A; Pepin, D; Mohan, R; Smith, S; Xia, T; Reinhardt, F; Chavarria, T; Hoefsmit, E; Pathania, S; Zhou, Y; Elias, K; Neel, B; Weinberg, R
Background The cornerstone of the existing treatment of high-grade serous ovarian cancer (HGSOC) is DNA-damaging chemotherapy; however, practically all patients eventually develop the progressive disease and the 5-year survival is only 40%. Immunotherapy would seem to be an attractive alternative treatment to chemotherapy, yet existing immunotherapies perform poorly in ovarian cancer, with only ~10% of patients responding to checkpoint-blockade. Why this is the case remains poorly understood and there is a pressing need to understand the underlying biology of immune evasion in ovarian cancer. Unfortunately, the preclinical tools required to explore the relationship between the types of DNA damage repair deficiencies and immune evasion have been lacking. Hence, we have modeled the biology of ovarian cancer using patient-relevant mutational landscapes in an immune-proficient, syngeneic-mouse model to help us identify the contribution of common driver mutations to the immune repertoire in the tumor microenvironment, and thus to responses of HGSOC tumors to immunotherapy. Methods We hypothesize that the immune composition and gene expression signatures of the resulting tumors will vary based on the combination of genetic alterations and the DNA repair proficiency of the transformed cells. To this end, we have engineered novel syngeneic mouse models from murine fallopian tube epithelium using CRISPR/Cas9 technology. These tumors capture the most common combinations of cooccurring mutations observed in patients. These models can identify the contribution of common driver mutations to the heterotypic interactions between cancer and stromal/immune compartments and examine how DNA repair proficiency contributes to immunogenicity. Results To validate the DNA repair proficiency of the transformed cells, we measured Rad51 nuclear focus formation after ionizing radiation (IR) and PARPinhibitor and DNA-damaging-agent sensitivity. The HR-deficient cell lines had significantly fewer Rad51 nuclear foci and were more sensitive to PARP-inhibition in comparison to HR-proficient cells. Initial immune /stromal analysis using flow cytometry, scRNAseq transcriptomic and immunofluorescence analysis revealed substantial differences in the myeloid and T-cell regulatory compartments between HR-proficient and-deficient primary and metastatic tumors and within the ascitic fluid. Preliminary results also suggest that inhibition of the DNA damage response, checkpoint kinase 1 in combination with immune checkpoint inhibitors, potentiates antitumor effects and augments cytotoxic T-cell infiltration. Conclusions These results reveal how common mutational drivers determine the microenvironment of the tumor and its response to treatment. Understanding the genetic basis of these complex cellular interactions will be critical to better tailor combinations of existing targeted treatments and immunotherapies in ovarian cancer to fight this devastating disease
EMBASE:629905285
ISSN: 2051-1426
CID: 4226682