Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Curr Protoc ; 3(7): e818, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37399289

ABSTRACT

Examination of DNA variation is central to understanding the function of mammalian cells, tissues, and whole bodies. Extraction of high-quality DNA from cells and tissues is necessary for innumerable different experiments. We present protocols for the extraction of DNA from both fresh samples and formalin-fixed tissue. Methods for extracting DNA have been standardized and streamlined over the past couple of decades and many extraction kits are available for a reasonable cost. In addition, many of the extraction procedures can also be automated for even higher throughput sample preparation. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: DNA isolation from whole blood, tissue, and cultured cells Alternate Protocol: DNA extraction using automated machines Basic Protocol 2: DNA isolation from saliva and buccal swabs Basic Protocol 3: DNA isolation from formalin-fixed, paraffin-embedded tissue.


Subject(s)
DNA , Formaldehyde , Animals , Tissue Fixation , DNA/genetics , Specimen Handling/methods , Genomics , Mammals/genetics
2.
Alzheimers Dement ; 19(2): 611-620, 2023 02.
Article in English | MEDLINE | ID: mdl-35490390

ABSTRACT

INTRODUCTION: Studies of cognitive impairment (CI) in Amish communities have identified sibships containing CI and cognitively unimpaired (CU) individuals. We hypothesize that CU individuals may carry protective alleles delaying age at onset (AAO) of CI. METHODS: A total of 1522 individuals screened for CI were genotyped. The outcome studied was AAO for CI individuals or age at last normal exam for CU individuals. Cox mixed-effects models examined association between age and single nucleotide variants (SNVs). RESULTS: Three SNVs were significantly associated (P < 5 × 10-8 ) with AAO on chromosomes 6 (rs14538074; hazard ratio [HR] = 3.35), 9 (rs534551495; HR = 2.82), and 17 (rs146729640; HR = 6.38). The chromosome 17 association was replicated in the independent National Institute on Aging Genetics Initiative for Late-Onset Alzheimer's Disease dataset. DISCUSSION: The replicated genome-wide significant association with AAO on chromosome 17 is located in the SHISA6 gene, which is involved in post-synaptic transmission in the hippocampus and is a biologically plausible candidate gene for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/genetics , Genome-Wide Association Study , Genotype , Cognitive Dysfunction/genetics , Polymorphism, Single Nucleotide
3.
medRxiv ; 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38168325

ABSTRACT

INTRODUCTION: Alzheimer disease (AD) remains a debilitating condition with limited treatments and additional therapeutic targets needed. Identifying AD protective genetic loci may identify new targets and accelerate identification of therapeutic treatments. We examined a founder population to identify loci associated with cognitive preservation into advanced age. METHODS: Genome-wide association and linkage analyses were performed on 946 examined and sampled Amish individuals, aged 76-95, who were either cognitively unimpaired (CU) or impaired (CI). RESULTS: 12 SNPs demonstrated suggestive association (P≤5×10-4) with cognitive preservation. Genetic linkage analyses identified >100 significant (LOD≥3.3) SNPs, some which overlapped with the association results. Only one locus on chromosome 2 retained significance across multiple analyses. DISCUSSION: A novel significant result for cognitive preservation on chromosome 2 includes the genes LRRTM4 and CTNNA2. Additionally, the lead SNP, rs1402906, impacts the POU3F2 transcription factor binding affinity, which regulates LRRTM4 and CTNNA2.

4.
Sci Rep ; 9(1): 13396, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31527715

ABSTRACT

Pancreatic ductal adenocarcinomas (PDAC) are deadly on account of the delay in diagnosis and dearth of effective treatment options for advanced disease. The insurmountable hurdle of targeting oncogene KRAS, the most prevalent genetic mutation in PDAC, has delayed the availability of targeted therapy for PDAC patients. An alternate approach is to target other tumour-exclusive effector proteins important in RAS signalling. The Family with Sequence Similarity 83 (FAM83) proteins are oncogenic, tumour-exclusive and function similarly to RAS, by driving the activation of PI3K and MAPK signalling. In this study we show that FAM83A expression is significantly elevated in human and murine pancreatic cancers and is essential for the growth and tumorigenesis of pancreatic cancer cells. Elevated FAM83A expression maintains essential MEK/ERK survival signalling, preventing cell death in pancreatic cancer cells. Moreover, we identified a positive feed-forward loop mediated by the MEK/ERK-activated AP-1 transcription factors, JUNB and FOSB, which is responsible for the elevated expression of oncogenic FAM83A. Our data indicates that targeting the MEK/ERK-FAM83A feed-forward loop opens up additional avenues for clinical therapy that bypass targeting of oncogenic KRAS in aggressive pancreatic cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Feedback, Physiological , Female , Humans , Mice , Mice, Nude , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Curr Protoc Hum Genet ; 92: 20.4.1-20.4.13, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28075484

ABSTRACT

The study of epigenetic properties of the human genome, including structural modifications of DNA and chromatin, has increased tremendously as mounting evidence has demonstrated how much epigenetics affects human gene expression. Buenrostro et al. have developed a rapid method, requiring low numbers of living cells as input, for examining chromatin accessibility across the epigenome, known as the assay for transposase-accessible chromatin using sequencing (ATAC-seq). The overall goal of this unit is to provide a thorough ATAC-seq data analysis plan, as well as describe how primary human blood samples can be processed for use in ATAC-seq. In addition, a number of quality control parameters are discussed to ensure the integrity and confidence in the ATAC-seq data. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Sequence Analysis, DNA/methods , Transposases/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Epigenesis, Genetic , Humans , Quality Control , Sequence Analysis, DNA/standards
6.
Clin Cancer Res ; 22(13): 3249-59, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26842237

ABSTRACT

PURPOSE: Recent data suggest that intrinsic subtype and immune cell infiltration may predict response to trastuzumab-based therapy. We studied the interaction between these factors, changes in immune signatures following brief exposure to trastuzumab, and achievement of pathologic complete response (pCR) to subsequent preoperative trastuzumab and chemotherapy in HER2-positive breast cancer. EXPERIMENTAL DESIGN: In patients enrolled on two multicenter trials (03-311 and 211B), tumor core biopsies were obtained at baseline and after brief exposure to single-agent trastuzumab or nab-paclitaxel. Gene expression profiles were assessed to assign PAM50 subtypes, measure immune cell activation, and were correlated with response. RESULTS: The pCR rate was significantly higher in HER2-enriched tumors in the Discovery, 03-311 (36%, P = 0.043) dataset, as compared with other subtypes, which validated in 211B (50%, P = 0.048). Significant increases in a signature of immune cell admixture (Immune Index) were observed only following brief exposure to trastuzumab in HER2-enriched tumors (Discovery/03-311, P = 0.05; Validation/211B, P = 0.02). Increased Immune Index was predictive of response after brief exposure (03-311, P = 0.03; 211B, P = 0.04), but not at baseline, in addition to increased expression of a CD4(+) follicular helper T-cell signature (03-311, P = 0.05; 211B, P = 0.04). Brief exposure to trastuzumab significantly increased gene expression of the T-cell marker PD-1 in HER2-enriched tumors (Discovery/03-311, P = 0.045) and PD-1 positivity by IHC (Validation/211B, P = 0.035). CONCLUSIONS: Correlations between pCR rates, increases in Immune Index and markers of T-cell activity following brief exposure to trastuzumab in HER2-enriched tumors provide novel insights into the interaction between tumor biology, antitumor immunity, and response to treatment, and suggest potential clinically useful biomarkers in HER2(+) breast cancers. Clin Cancer Res; 22(13); 3249-59. ©2016 AACR.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/drug therapy , Immunity, Innate/immunology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Trastuzumab/therapeutic use , Albumins/therapeutic use , B-Lymphocytes/immunology , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Lymphocyte Activation/immunology , Macrophages/immunology , Middle Aged , Neoadjuvant Therapy , Paclitaxel/therapeutic use , Programmed Cell Death 1 Receptor/biosynthesis , Treatment Outcome
7.
Int J Cancer ; 138(3): 747-57, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26284485

ABSTRACT

To best define biomarkers of response, and to shed insight on mechanism of action of certain clinically important agents for early breast cancer, we used a brief-exposure paradigm in the preoperative setting to study transcriptional changes in patient tumors that occur with one dose of therapy prior to combination chemotherapy. Tumor biopsies from breast cancer patients enrolled in two preoperative clinical trials were obtained at baseline and after one dose of bevacizumab (HER2-negative), trastuzumab (HER2-positive) or nab-paclitaxel, followed by treatment with combination chemo-biologic therapy. RNA-Sequencing based PAM50 subtyping at baseline of 46 HER2-negative patients revealed a strong association between the basal-like subtype and pathologic complete response (pCR) to chemotherapy plus bevacizumab (p ≤ 0.0027), but did not provide sufficient specificity to predict response. However, a single dose of bevacizumab resulted in down-regulation of a well-characterized TGF-ß activity signature in every single breast tumor that achieved pCR (p ≤ 0.004). The TGF-ß signature was confirmed to be a tumor-specific read-out of the canonical TGF-ß pathway using pSMAD2 (p ≤ 0.04), with predictive power unique to brief-exposure to bevacizumab (p ≤ 0.016), but not trastuzumab or nab-paclitaxel. Down-regulation of TGF-ß activity was associated with reduction in tumor hypoxia by transcription and protein levels, suggesting therapy-induced disruption of an autocrine-loop between tumor stroma and malignant cells. Modulation of the TGF-ß pathway upon brief-exposure to bevacizumab may provide an early functional readout of pCR to preoperative anti-angiogenic therapy in HER2-negative breast cancer, thus providing additional avenues for exploration in both preclinical and clinical settings with these agents.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/analysis , Transforming Growth Factor beta/physiology , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Hypoxia , Female , Humans , Sequence Analysis, RNA , Signal Transduction/physiology
8.
Breast Cancer Res Treat ; 150(2): 321-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25749757

ABSTRACT

Breast cancer is a major health problem affecting millions of women worldwide. Over 200,000 new cases are diagnosed annually in the USA, with approximately 40,000 of these cases resulting in death. HER2-positive (HER2+) breast tumors, representing 20-30 % of early-stage breast cancer diagnoses, are characterized by the amplification of the HER2 gene. However, the critical genes and pathways that become affected by HER2 amplification in humans are yet to be specifically identified. Furthermore, it is yet to be determined if HER2 amplification also affects the expression of long intervening non-coding (linc)RNAs, which are involved in the epigenetic regulation of gene expression. We examined changes in gene expression by next generation RNA sequencing in human tumors pre- and post- HER2 inhibition by trastuzumab in vivo, and changes in gene expression in response to HER2 knock down in cell culture models. We integrated our results with gene expression analysis of HER2+ tumors vs matched normal tissue from The Cancer Genome Atlas. The integrative analyses of these datasets led to the identification of a small set of mRNAs, and the associated biological pathways that become deregulated by HER2 amplification. Furthermore, our analyses identified three lincRNAs that become deregulated in response to HER2 amplification both in vitro and in vivo. Our results should provide the foundation for functional studies of these candidate mRNAs and lincRNAs to further our understanding of how HER2 amplification results in tumorigenesis. Also, the identified lincRNAs could potentially open the door for future RNA-based biomarkers and therapeutics in HER2+ breast cancer.


Subject(s)
Breast Neoplasms/metabolism , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Transcriptome , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Cell Line, Tumor , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Sequence Analysis, RNA , Signal Transduction , Trastuzumab/pharmacology
9.
Mol Cancer Res ; 12(8): 1156-65, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24736947

ABSTRACT

UNLABELLED: FAM83B (family with sequence similarity 83, member B) was recently identified as a novel oncogene involved in activating CRAF/MAPK signaling and driving epithelial cell transformation. FAM83B is one of eight members of a protein family (FAM83) characterized by a highly conserved domain of unknown function (DUF1669), which is necessary and sufficient to drive transformation. Here, it is demonstrated that additional FAM83 members also exhibit oncogenic properties and have significantly elevated levels of expression in multiple human tumor types using a TissueScan Cancer Survey Panel PCR array and database mining. Furthermore, modeling the observed tumor expression of FAM83A, FAM83C, FAM83D, or FAM83E promoted human mammary epithelial cell (HMEC) transformation, which correlated with the ability of each FAM83 member to bind CRAF (RAF1) and promote CRAF membrane localization. Conversely, ablation of FAM83A or FAM83D from breast cancer cells resulted in diminished MAPK signaling with marked suppression of growth in vitro and tumorigenicity in vivo. Importantly, each FAM83 member was determined to be elevated in at least one of 17 distinct tumor types examined, with FAM83A, FAM83B, and FAM83D most frequently overexpressed in several diverse tissue types. Finally, evidence suggests that elevated expression of FAM83 members is associated with elevated tumor grade and decreased overall survival. IMPLICATIONS: FAM83 proteins represent a novel family of oncogenes suitable for the development of cancer therapies aimed at suppressing MAPK signaling.


Subject(s)
Breast Neoplasms/genetics , Mitogen-Activated Protein Kinases/genetics , Neoplasm Proteins/genetics , Signal Transduction/genetics , Animals , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Epithelial Cells/metabolism , Female , Humans , Mammary Glands, Human/metabolism , Mice , Mice, Nude , Oncogenes/genetics
10.
Oncotarget ; 4(5): 729-38, 2013 May.
Article in English | MEDLINE | ID: mdl-23676467

ABSTRACT

Therapies targeting MAPK and AKT/mTOR signaling are currently being evaluated in clinical trials for several tumor types. However, recent studies suggest that these therapies may be limited due to acquired cancer cell resistance and a small therapeutic index between normal and cancer cells. The identification of novel proteins that are involved in MAPK or AKT/mTOR signaling and differentially expressed between normal and cancer cells will provide mechanistically distinct therapeutic targets with the potential to inhibit these key cancer-associated pathways. We recently identified FAM83B as a novel, previously uncharacterized oncogene capable of hyperactivating MAPK and mTOR signaling and driving the tumorigenicity of immortalized human mammary epithelial cells (HMEC). We show here that elevated FAM83B expression also activates the PI3K/AKT signaling pathway and confers a decreased sensitivity to PI3K, AKT, and mTOR inhibitors. FAM83B co-precipitated with the p85α and p110α subunits of PI3K, as well as AKT, and increased p110α and AKT membrane localization, consistent with elevated PI3K/AKT signaling. In tumor-derived cells harboring elevated FAM83B expression, ablation of FAM83B decreased p110α and AKT membrane localization, suppressed AKT phosphorylation, and diminished proliferation, AIG, and tumorigenicity in vivo. We propose that the level of FAM83B expression may be an important factor to consider when combined therapies targeting MAPK and AKT/mTOR signaling are used. Moreover, the identification of FAM83B as a novel oncogene and its integral involvement in activating PI3K/AKT and MAPK provides a foundation for future therapies aimed at targeting FAM83B in order to suppress the growth of PI3K/AKT- and MAPK-driven cancers.


Subject(s)
Colonic Neoplasms/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Drug Resistance, Neoplasm , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Knockout Techniques , HCT116 Cells , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Transplantation , TOR Serine-Threonine Kinases/metabolism
11.
J Clin Invest ; 122(9): 3197-210, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22886302

ABSTRACT

Aberrant regulation of growth signaling is a hallmark of cancer development that often occurs through the constitutive activation of growth factor receptors or their downstream effectors. Using validation-based insertional mutagenesis (VBIM), we identified family with sequence similarity 83, member B (FAM83B), based on its ability to substitute for RAS in the transformation of immortalized human mammary epithelial cells (HMECs). We found that FAM83B coprecipitated with a downstream effector of RAS, CRAF. Binding of FAM83B with CRAF disrupted CRAF/14-3-3 interactions and increased CRAF membrane localization, resulting in elevated MAPK and mammalian target of rapamycin (mTOR) signaling. Ablation of FAM83B inhibited the proliferation and malignant phenotype of tumor-derived cells or RAS-transformed HMECs, implicating FAM83B as a key intermediary in EGFR/RAS/MAPK signaling. Analysis of human tumor specimens revealed that FAM83B expression was significantly elevated in cancer and was associated with specific cancer subtypes, increased tumor grade, and decreased overall survival. Cumulatively, these results suggest that FAM83B is an oncogene and potentially represents a new target for therapeutic intervention.


Subject(s)
Cell Transformation, Neoplastic/metabolism , ErbB Receptors/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gene Expression , Gene Knockdown Techniques , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/metabolism , Quinazolines/pharmacology , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transcriptome
12.
PLoS One ; 5(2): e9152, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20161778

ABSTRACT

BACKGROUND: Grb2-associated binding (Gab) adapter proteins play major roles in coordinating signaling downstream of hematopoietic cytokine receptors. In hematopoietic cells, Gab2 can modulate phosphatidylinositol-3 kinase and mitogen associated protein kinase activities and regulate the long-term multilineage competitive repopulating activity of hematopoietic stem cells (HSCs). Gab2 may also act in a linear pathway upstream or downstream of signal transducer and activator of transcription-5 (STAT5), a major positive regulator of HSC function. Therefore, we aimed to determine whether Gab2 and STAT5 function in hematopoiesis in a redundant or non-redundant manner. METHODOLOGY/PRINCIPAL FINDINGS: To do this we generated Gab2 mutant mice with heterozygous and homozygous deletions of STAT5. In heterozygous STAT5 mutant mice, deficiencies in HSC/multipotent progenitors were reflected by decreased long-term repopulating activity. This reduction in repopulation function was mirrored in the reduced growth response to early-acting cytokines from sorted double mutant c-Kit(+)Lin(-)Sca-1(+) (KLS) cells. Importantly, in non-ablated newborn mice, the host steady-state engraftment ability was impaired by loss of Gab2 in heterozygous STAT5 mutant background. Fetal liver cells isolated from homozygous STAT5 mutant mice lacking Gab2 showed significant reduction in HSC number (KLS CD150(+)CD48(-)), reduced HSC survival, and dramatic loss of self-renewal potential as measured by serial transplantation. CONCLUSIONS/SIGNIFICANCE: These data demonstrate new functions for Gab2 in hematopoiesis in a manner that is non-redundant with STAT5. Furthermore, important synergy between STAT5 and Gab2 was observed in HSC self-renewal, which might be exploited to optimize stem cell-based therapeutics.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/metabolism , Phosphoproteins/deficiency , STAT5 Transcription Factor/deficiency , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Cell Lineage , Cell Proliferation , Cell Survival , Colony-Forming Units Assay , Female , Flow Cytometry , Genotype , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Liver/cytology , Liver/embryology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , STAT5 Transcription Factor/genetics
13.
Blood ; 115(7): 1416-24, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20008792

ABSTRACT

Phosphorylated signal transducer and activator of transcription 5 (STAT5) is a biomarker and potential molecular target for hematologic malignancies. We have shown previously that lethal myeloproliferative disease (MPD) in mice mediated by persistently activated STAT5 (STAT5a(S711F)) requires the N-domain, but the mechanism was not defined. We now demonstrate by retrovirally complementing STAT5ab(null/null) primary mast cells that relative to wild-type STAT5a, STAT5a lacking the N-domain (STAT5aDeltaN) ineffectively protected against cytokine withdrawal-induced cell death. Both STAT5a and STAT5aDeltaN bound to a site in the bcl-2 gene and both bound near the microRNA 15b/16 cluster. However, only STAT5a could effectively induce bcl-2 mRNA and reciprocally suppress miR15b/16 leading to maintained bcl-2 protein levels. After retroviral complementation of STAT5ab(null/null) fetal liver cells and transplantation, persistently active STAT5a(S711F) lacking the N-domain (STAT5aDeltaN(S711F)) was insufficient to protect c-Kit(+)Lin(-)Sca-1(+) (KLS) cells from apoptosis and unable to induce bcl-2 expression, whereas STAT5a(S711F) caused robust KLS cell expansion, induction of bcl-2, and lethal MPD. Severe attenuation of MPD by STAT5aDeltaN(S711F) was reversed by H2k/bcl-2 transgenic expression. Overall, these studies define N-domain-dependent survival signaling as an Achilles heel of persistent STAT5 activation and highlight the potential therapeutic importance of targeting STAT5 N-domain-mediated regulation of bcl-2 family members.


Subject(s)
MicroRNAs/metabolism , Myeloproliferative Disorders/physiopathology , Proto-Oncogene Proteins c-bcl-2/genetics , STAT5 Transcription Factor/genetics , Animals , Cell Survival/physiology , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Introns/physiology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphinans/metabolism , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Protein Structure, Tertiary , RNA Processing, Post-Transcriptional/physiology , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...