Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
NAR Cancer ; 4(2): zcac014, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35475145

ABSTRACT

We created the PDX Network (PDXNet) portal (https://portal.pdxnetwork.org/) to centralize access to the National Cancer Institute-funded PDXNet consortium resources, to facilitate collaboration among researchers and to make these data easily available for research. The portal includes sections for resources, analysis results, metrics for PDXNet activities, data processing protocols and training materials for processing PDX data. Currently, the portal contains PDXNet model information and data resources from 334 new models across 33 cancer types. Tissue samples of these models were deposited in the NCI's Patient-Derived Model Repository (PDMR) for public access. These models have 2134 associated sequencing files from 873 samples across 308 patients, which are hosted on the Cancer Genomics Cloud powered by Seven Bridges and the NCI Cancer Data Service for long-term storage and access with dbGaP permissions. The portal includes results from freely available, robust, validated and standardized analysis workflows on PDXNet sequencing files and PDMR data (3857 samples from 629 patients across 85 disease types). The PDXNet portal is continuously updated with new data and is of significant utility to the cancer research community as it provides a centralized location for PDXNet resources, which support multi-agent treatment studies, determination of sensitivity and resistance mechanisms, and preclinical trials.

2.
J Natl Med Assoc ; 112(3): 308-314, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32553457

ABSTRACT

The National Cancer Institute's (NCI) Center to Reduce Cancer Health Disparities (CRCHD) was established in 2001 with the purpose of confronting and eliminating cancer health disparities, while increasing workforce diversity in cancer research. Over the last two decades, CRCHD has generated a broad range of research, training, and community outreach activities to address these overarching goals through a variety of programs including the Continuing Umbrella of Research Experiences (CURE), Partnerships to Advance Cancer Health Equity (PACHE), Special Populations Networks (SPN), Community Networks Program (CNP), CNP Centers (CNPC), and Patient Navigation Research Program (PNRP). CRCHD, through its CURE and now its Intramural CURE (iCURE) programs, has been fully dedicated to training the next generation of competitive researchers from backgrounds typically underrepresented in the cancer and cancer health disparities research fields. Today, CRCHD leads NCI's efforts in supporting research training and career development experiences beginning as early as middle school and continuing through to tenured track appointments. CRCHD has also developed a robust basic research focus in cancer disparities, which has recently expanded into translational disparities research and the generation of novel, authenticated animal models appropriate for advancing disparities research investigations. Additionally, CRCHD has fostered an integrated networks infrastructure to complement and support its disparities research and diversity training efforts, as well as provide cancer education and outreach among racially and ethnically diverse and medically underserved communities. Moving forward, the CRCHD will continue its steadfast efforts to move us closer to the day when diversity is a given and disparities no longer exist.


Subject(s)
Health Equity , Health Promotion , Health Status Disparities , Health Workforce , Neoplasms/ethnology , Health Personnel , Healthcare Disparities , Humans , National Cancer Institute (U.S.) , National Institutes of Health (U.S.) , United States
3.
Clin Cancer Res ; 24(21): 5471-5481, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30012562

ABSTRACT

Purpose: Men of African ancestry experience an excessive prostate cancer mortality that could be related to an aggressive tumor biology. We previously described an immune-inflammation signature in prostate tumors of African-American (AA) patients. Here, we further deconstructed this signature and investigated its relationships with tumor biology, survival, and a common germline variant in the IFNλ4 (IFNL4) gene.Experimental Design: We analyzed gene expression in prostate tissue datasets and performed genotype and survival analyses. We also overexpressed IFNL4 in human prostate cancer cells.Results: We found that a distinct interferon (IFN) signature that is analogous to the previously described "IFN-related DNA damage resistance signature" (IRDS) occurs in prostate tumors. Evaluation of two independent patient cohorts revealed that IRDS is detected about twice as often in prostate tumors of AA than European-American men. Furthermore, analysis in TCGA showed an association of increased IRDS in prostate tumors with decreased disease-free survival. To explain these observations, we assessed whether IRDS is associated with an IFNL4 germline variant (rs368234815-ΔG) that controls production of IFNλ4, a type III IFN, and is most common in individuals of African ancestry. We show that the IFNL4 rs368234815-ΔG allele was significantly associated with IRDS in prostate tumors and overall survival of AA patients. Moreover, IFNL4 overexpression induced IRDS in three human prostate cancer cell lines.Conclusions: Our study links a germline variant that controls production of IFNλ4 to the occurrence of a clinically relevant IFN signature in prostate tumors that may predominantly affect men of African ancestry. Clin Cancer Res; 24(21); 5471-81. ©2018 AACR.


Subject(s)
Alleles , Interleukins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Sequence Deletion , Biomarkers , Gene Expression Profiling , Genotype , Germ-Line Mutation , Humans , Interferons/genetics , Interferons/metabolism , Interleukins/metabolism , Male , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Recurrence , Transcriptome , Tryptophan/metabolism
4.
Cancer Res ; 76(5): 1055-1065, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26719530

ABSTRACT

Smokers develop metastatic prostate cancer more frequently than nonsmokers, suggesting that a tobacco-derived factor is driving metastatic progression. To identify smoking-induced alterations in human prostate cancer, we analyzed gene and protein expression patterns in tumors collected from current, past, and never smokers. By this route, we elucidated a distinct pattern of molecular alterations characterized by an immune and inflammation signature in tumors from current smokers that were either attenuated or absent in past and never smokers. Specifically, this signature included elevated immunoglobulin expression by tumor-infiltrating B cells, NF-κB activation, and increased chemokine expression. In an alternate approach to characterize smoking-induced oncogenic alterations, we also explored the effects of nicotine in human prostate cancer cells and prostate cancer-prone TRAMP mice. These investigations showed that nicotine increased glutamine consumption and invasiveness of cancer cells in vitro and accelerated metastatic progression in tumor-bearing TRAMP mice. Overall, our findings suggest that nicotine is sufficient to induce a phenotype resembling the epidemiology of smoking-associated prostate cancer progression, illuminating a novel candidate driver underlying metastatic prostate cancer in current smokers.


Subject(s)
Inflammation/metabolism , Prostatic Neoplasms/immunology , Smoking/adverse effects , Transcriptome , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Immunoglobulins/genetics , Interleukin-8/blood , Male , Mice , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Nicotine/pharmacology , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism
5.
Carcinogenesis ; 35(9): 2074-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24858205

ABSTRACT

Aberrant expression of subgroup k human endogenous retroviruses (HERV-K) has been observed in prostate cancer. This subgroup is unique because it encodes sequences in the human genome containing open reading frames for near intact retroviruses. We hypothesized that HERV-K reactivation could serve as a non-invasive early disease detection marker for prostate cancer. We evaluated HERV-K gag messenger RNA (mRNA) expression in blood samples of African-American and European-American men using a case-control design via quantitative real-time PCR. Additionally, we examined HERV-K envelope protein expression in prostate tumors by immunohistochemistry. HERV-K envelope protein was commonly upregulated in prostate tumors, but more so in tumors of African-American than European-American patients (61% versus 40%, P < 0.01). Examining HERV-K gag expression in peripheral blood mononuclear cells (PBMC) from 294 cases and 135 healthy men, we found that the abundance of HERV-K gag message was significantly higher in cases than controls and was associated with increased plasma interferon-γ. Men with gag expression in the highest quartile had >12-fold increased odds {odds ratio = 12.87 [95% confidence interval 6.3-26.25]} of being diagnosed with prostate cancer than those in the lowest quartile. Moreover, our results showed that HERV-K expression may perform better as a disease biomarker in older than younger men (whereas the sensitivity of prostate-specific antigen (PSA) testing decreases with age) and in men with a smoking history compared with never smokers. Combining non-invasive HERV-K testing with PSA testing may improve the efficacy of prostate cancer detection specifically among older men and smokers who tend to develop a more aggressive disease.


Subject(s)
Adenocarcinoma/blood , Gene Products, gag/blood , Leukocytes, Mononuclear/metabolism , Prostatic Neoplasms/blood , Smoking/blood , Adenocarcinoma/diagnosis , Adenocarcinoma/virology , Chemokine CXCL10/blood , Endogenous Retroviruses/enzymology , Gene Expression , Humans , Interferon-gamma/blood , Leukocytes, Mononuclear/virology , Male , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/virology , RNA, Messenger/blood , RNA, Messenger/genetics , Risk Factors
6.
J Clin Invest ; 124(1): 398-412, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316975

ABSTRACT

Metabolic profiling of cancer cells has recently been established as a promising tool for the development of therapies and identification of cancer biomarkers. Here we characterized the metabolomic profile of human breast tumors and uncovered intrinsic metabolite signatures in these tumors using an untargeted discovery approach and validation of key metabolites. The oncometabolite 2-hydroxyglutarate (2HG) accumulated at high levels in a subset of tumors and human breast cancer cell lines. We discovered an association between increased 2HG levels and MYC pathway activation in breast cancer, and further corroborated this relationship using MYC overexpression and knockdown in human mammary epithelial and breast cancer cells. Further analyses revealed globally increased DNA methylation in 2HG-high tumors and identified a tumor subtype with high tissue 2HG and a distinct DNA methylation pattern that was associated with poor prognosis and occurred with higher frequency in African-American patients. Tumors of this subtype had a stem cell-like transcriptional signature and tended to overexpress glutaminase, suggestive of a functional relationship between glutamine and 2HG metabolism in breast cancer. Accordingly, 13C-labeled glutamine was incorporated into 2HG in cells with aberrant 2HG accumulation, whereas pharmacologic and siRNA-mediated glutaminase inhibition reduced 2HG levels. Our findings implicate 2HG as a candidate breast cancer oncometabolite associated with MYC activation and poor prognosis.


Subject(s)
Breast Neoplasms/metabolism , Glutarates/metabolism , Proto-Oncogene Proteins c-myc/physiology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/mortality , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glutamine/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , MCF-7 Cells , Metabolome , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Prognosis , RNA, Small Interfering/genetics , Receptors, Estrogen/metabolism , Survival Analysis , Transcriptome , Wnt Signaling Pathway
7.
PLoS One ; 8(3): e57361, 2013.
Article in English | MEDLINE | ID: mdl-23469190

ABSTRACT

BACKGROUND: A study from Scotland reported that the p53 mutation frequency in breast tumors is associated with socio-economic deprivation. METHODS: We analyzed the association of the tumor p53 mutational status with tumor characteristics, education, and self-reported annual household income (HI) among 173 breast cancer patients from the greater Baltimore area, United States. RESULTS: p53 mutational frequency was significantly associated with HI. Patients with < $15,000 HI had the highest p53 mutation frequency (21%), followed by the income group between $15,000 and $60,000 (18%), while those above $60,000 HI had the fewest mutations (5%). When dichotomized at $60,000, 26 out of 135 patients in the low income category had acquired a p53 mutation, while only 2 out of 38 with a high income carried a mutation (P < 0.05). In the adjusted logistic regression analysis with 3 income categories (trend test), the association between HI and p53 mutational status was independent of tumor characteristics, age, race/ethnicity, tobacco smoking and body mass. Further analyses revealed that HI may impact the p53 mutational frequency preferentially in patients who develop an estrogen receptor (ER)-negative disease. Within this group, 42% of the low income patients (< $15,000 HI) carried a mutation, followed by the middle income group (21%), while those above $60,000 HI did not carry mutations (Ptrend < 0.05). CONCLUSIONS: HI is associated with the p53 mutational frequency in patients who develop an ER-negative disease. Furthermore, high income patients may acquire fewer p53 mutations than other patients, suggesting that lifetime exposures associated with socio-economic status may impact breast cancer biology.


Subject(s)
Breast Neoplasms/genetics , Income/statistics & numerical data , Mutation Rate , Tumor Suppressor Protein p53/genetics , Adult , Aged , Baltimore/epidemiology , Black People , Body Mass Index , Breast Neoplasms/ethnology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Educational Status , Female , Humans , Middle Aged , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Regression Analysis , Risk Factors , Smoking , Survival Analysis , White People
8.
Carcinogenesis ; 32(8): 1107-21, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21464040

ABSTRACT

Cancer incidence and mortality rates show great variations across nations and between population groups. These variations are largely explained by differences in age distribution, diet and lifestyle, access to health care, cultural barriers and exposure to carcinogens and pathogens. Cancers caused by infections are significantly more common in developing than developed countries, and they overproportionally affect immigrant populations in the USA and other countries. The global pattern of cancer is not stagnant. Instead, it is dynamic because of fluctuations in the age distribution of populations, improvements in cancer prevention and early detection in affluent countries and rapid changes in diet and lifestyle in parts of the world. For example, increased smoking rates have caused tobacco-induced cancers to rise in various Asian countries, whereas reduced smoking rates have caused these cancers to plateau or even begin to decline in Western Europe and North America. Some population groups experience a disproportionally high cancer burden. In the USA and the Caribbean, cancer incidence and mortality rates are excessively high in populations of African ancestry when compared with other population groups. The causes of this disparity are multifaceted and may include tumor biological and genetic factors and their interaction with the environment. In this review, we will discuss the magnitude and causes of global cancer health disparities and will, with a focus on African-Americans and selected cancer sites, evaluate the evidence that genetic and tumor biological factors contribute to existing cancer incidence and outcome differences among population groups in the USA.


Subject(s)
Environment , Genes/physiology , Healthcare Disparities , Neoplasms/physiopathology , Global Health , Humans
9.
Cancer Res ; 68(15): 6162-70, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676839

ABSTRACT

MicroRNAs are small noncoding RNAs that regulate the expression of protein-coding genes. To evaluate the involvement of microRNAs in prostate cancer, we determined genome-wide expression of microRNAs and mRNAs in 60 primary prostate tumors and 16 nontumor prostate tissues. The mRNA analysis revealed that key components of microRNA processing and several microRNA host genes, e.g., MCM7 and C9orf5, were significantly up-regulated in prostate tumors. Consistent with these findings, tumors expressed the miR-106b-25 cluster, which maps to intron 13 of MCM7, and miR-32, which maps to intron 14 of C9orf5, at significantly higher levels than nontumor prostate. The expression levels of other microRNAs, including a number of miR-106b-25 cluster homologues, were also altered in prostate tumors. Additional differences in microRNA abundance were found between organ-confined tumors and those with extraprostatic disease extension. Lastly, we found evidence that some microRNAs are androgen-regulated and that tumor microRNAs influence transcript abundance of protein-coding target genes in the cancerous prostate. In cell culture, E2F1 and p21/WAF1 were identified as targets of miR-106b, Bim of miR-32, and exportin-6 and protein tyrosine kinase 9 of miR-1. In summary, microRNA expression becomes altered with the development and progression of prostate cancer. Some of these microRNAs regulate the expression of cancer-related genes in prostate cancer cells.


Subject(s)
Gene Expression Profiling , Genomics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , Aged , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Prostatic Neoplasms/enzymology , Ribonuclease III/metabolism , Up-Regulation
10.
Prostate ; 68(11): 1152-64, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18459106

ABSTRACT

BACKGROUND: Perineural invasion (PNI) is the dominant pathway for local invasion in prostate cancer. To date, only few studies have investigated the molecular differences between prostate tumors with PNI and those without it. METHODS: To evaluate the involvement of both microRNAs and protein-coding genes in PNI, we determined their genome-wide expression with a custom microRNA microarray and Affymetrix GeneChips in 50 prostate adenocarcinomas with PNI and 7 without it. In situ hybridization (ISH) and immunohistochemistry was used to validate candidate genes. RESULTS: Unsupervised classification of the 57 adenocarcinomas revealed two clusters of tumors with distinct global microRNA expression. One cluster contained all non-PNI tumors and a subgroup of PNI tumors. Significance analysis of microarray data yielded a list of microRNAs associated with PNI. At a false discovery rate (FDR)<10%, 19 microRNAs were higher expressed in PNI tumors than in non-PNI tumors. The most differently expressed microRNA was miR-224. ISH showed that this microRNA is expressed by perineural cancer cells. The analysis of protein-coding genes identified 34 transcripts that were differently expressed by PNI status (FDR<10%). These transcripts were down-regulated in PNI tumors. Many of those encoded metallothioneins and proteins with mitochondrial localization and involvement in cell metabolism. Consistent with the microarray data, perineural cancer cells tended to have lower metallothionein expression by immunohistochemistry than nonperineural cancer cells. CONCLUSIONS: Although preliminary, our findings suggest that alterations in microRNA expression, mitochondrial function, and cell metabolism occur at the transition from a noninvasive prostate tumor to a tumor with PNI.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Cell Differentiation/physiology , Cell Lineage/physiology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Humans , Male , Metallothionein/genetics , Mitochondria/physiology , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Peripheral Nerves/pathology , Receptors, Virus/genetics
11.
Cancer Res ; 68(3): 927-36, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18245496

ABSTRACT

The incidence and mortality rates of prostate cancer are significantly higher in African-American men when compared with European-American men. We tested the hypothesis that differences in tumor biology contribute to this survival health disparity. Using microarray technology, we obtained gene expression profiles of primary prostate tumors resected from 33 African-American and 36 European-American patients. These tumors were matched on clinical variables. We also evaluated 18 nontumor prostate tissues from seven African-American and 11 European-American patients. The resulting datasets were analyzed for expression differences on the gene and pathway level comparing African-American with European-American patients. Our analysis revealed a significant number of genes, e.g., 162 transcripts at a false-discovery rate of

Subject(s)
Black People , Black or African American , Prostatic Neoplasms/ethnology , White People , Gene Expression Profiling , Humans , Male , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
12.
Exp Cell Res ; 312(15): 2735-44, 2006 Sep 10.
Article in English | MEDLINE | ID: mdl-16887119

ABSTRACT

Jak2 is a member of the Janus family of tyrosine kinases and is involved in cytokine signaling. As a part of a study to determine biological functions of Jak2, we used molecular modeling to identify W1038 as a residue that is critical for tyrosine kinase function. Mutation of W1038, in tandem with E1046, generates a dominant-negative form of the Jak2 protein. Mice that were engineered to express two copies of this dominant-negative Jak2 protein died in utero. Additionally, heterozygous mice expressing Jak2 with kinase activity that is moderately reduced when compared to wild-type activity appear phenotypically normal. Collectively, these data suggest that Jak2 kinase activity is essential for normal mammalian development.


Subject(s)
Embryonic Development , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Alleles , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cells, Cultured , Dimerization , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Hematopoiesis/physiology , Janus Kinase 2 , Mice , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Phenotype , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism
13.
Cell Biochem Biophys ; 44(2): 213-22, 2006.
Article in English | MEDLINE | ID: mdl-16456223

ABSTRACT

Jak2 is a nonreceptor tyrosine kinase that is essential for proper animal development and physiology. It is activated by ligand-occupied cell-surface receptors. Once activated, it then tyrosine phosphorylates the latent cytoplasmic transcription factors, termed the signal transducers and activators of transcription (STAT) proteins. Thus Jak2 is viewed as a classic mediator of ligand-dependent signal transduction. Recent studies, however, suggest that Jak2 may mediate cellular gene expression outside of the classically defined, ligand-activated, Jak/STAT-signaling paradigm. Here we review these studies, provide additional data, and discuss whether Jak2 is a mediator of ligand-independent gene transcription, and, in turn, whether our current understanding of the Jak/STAT signaling paradigm should be modified to incorporate these observations.


Subject(s)
Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Signal Transduction , Animals , Gene Expression Regulation , Humans , Janus Kinase 2 , Ligands , Signal Transduction/genetics , Transcription, Genetic
14.
Vascul Pharmacol ; 43(5): 336-45, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16257270

ABSTRACT

In addition to its role as a vasoconstrictor, angiotensin II also acts as a potent growth factor by activating several tyrosine kinases, including Jak2. Interestingly, Jak2 has been linked to similar cardiovascular pathologies as have been previously linked to the renin-angiotensin system. Identifying the downstream targets of Jak2 via the AT(1) receptor may therefore elucidate its role in the progression of various pathologies. Previously, microarray analysis from our laboratory identified the Type 1 inositol 1,4,5 trisphosphate (IP(3)) receptor as a potential target of Jak2 following chronic stimulation by angiotensin II. Therefore, we hypothesized that Jak2 regulates IP(3) receptor expression in response to angiotensin II. To test this hypothesis, rat aortic smooth muscle (RASM) cells over-expressing a dominant negative (DN) Jak2 protein were used. The Jak2-dependent signaling in these cells is reduced approximately 90% when compared to RASM control cells. Analysis of protein expression showed that the IP(3) receptor was degraded approximately 2-fold (P<0.05) in cells lacking functional Jak2 within 1 h of treatment by angiotensin II. Notably, degradation of the IP(3) receptor was reversible since protein levels were restored to normal following 2 h of recovery from angiotensin II. To eliminate the possibility of clonal artifact in the DN cells, wild type RASM cells were treated with the Jak2 pharmacological inhibitor, AG490. We found that angiotensin II treatment degraded IP(3) receptor in AG490-treated cells, but not in the vehicle controls. Treatment with lactacystin, a proteasome inhibitor, completely blocked angiotensin II-mediated degradation of IP(3) receptor, thereby suggesting that the degradation occurs through a proteasome-dependent mechanism. Moreover, the degradation of IP(3) receptor in DN cells correlated with a significant loss of intracellular calcium mobilization when treated with angiotensin II (DN 27.4+/-1.1% vs. WT 42.2+/-4.7%; n=5, P=0.002). We next examined through what mechanism Jak2 regulates the IP(3) receptor. When wild type RASM cells were treated with PP2, an Src-family inhibitor, IP(3) receptor expression was markedly reduced. Since previous data show that Fyn, a downstream target of Jak2, is able to phosphorylate the IP(3) receptor at Tyr 353, we believe our data suggest that Jak2 prevents the angiotensin II-mediated IP(3) receptor degradation through the activation of Fyn. In conclusion, these data suggest that Jak2 has a protective role in maintaining IP(3) receptor expression, potentially through activation of Fyn and subsequent phosphorylation of the IP(3) receptor.


Subject(s)
Angiotensin II/antagonists & inhibitors , Calcium Channels/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Blotting, Western , Calcium/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Inositol 1,4,5-Trisphosphate Receptors , Janus Kinase 2 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Tyrphostins/pharmacology
15.
Cell Biochem Biophys ; 41(2): 207-32, 2004.
Article in English | MEDLINE | ID: mdl-15475610

ABSTRACT

Discovered roughly 10 yr ago, Jak2 tyrosine kinase has emerged as a critical molecule in mammalian development, physiology, and disease. Here, we review the early history of Jak2 and its role in health and disease. We will also review its critical role in mediating cytokine-dependent signal transduction. Additionally, more recent work demonstrating the importance of Jak2 in G protein-coupled receptor and tyrosine kinase growth factor receptor signal transduction will be discussed. The cellular and biochemical mechanisms by which Jak2 tyrosine kinase is activated and regulated within the cell also will be reviewed. Finally, structure-function and pharmacological-based studies that identified structural motifs and amino acids within Jak2 that are critical for its function will be examined. By reviewing the biology of Jak2 tyrosine kinase at the molecular, cellular, and physiological levels, we hope to advance the understanding of how a single gene can have such a profound impact on development, physiology, and disease.


Subject(s)
Janus Kinase 2/physiology , Transcription, Genetic , Amino Acid Motifs , Animals , Cell Nucleus/metabolism , Cytokines/metabolism , Gene Expression Regulation, Enzymologic , Humans , Janus Kinase 2/metabolism , Ligands , Mice , Models, Biological , Phosphorylation , Protein Structure, Tertiary , Signal Transduction , Structure-Activity Relationship
16.
Am J Physiol Cell Physiol ; 287(4): C981-91, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15189810

ABSTRACT

Mice lacking a functional Janus kinase 2 (JAK2) allele die embryonically, indicating the mandatory role of JAK2 in basic developmental cellular transcription. Currently, however, the downstream target genes of JAK2 are largely unknown. Here, in vitro conditions were created using a cell line lacking JAK2 expression. Microarray analysis was then used to identify genes that are differentially expressed as a result of the presence, or absence, of JAK2. The data identified 621 JAK2-dependent genes as having at least a twofold change in expression. Surprisingly, these genes did not require ligand-dependent activation of JAK2 but merely its expression in the cell. Thirty-one of these genes were found to have a greater than sevenfold change in expression levels, and a subset of these were further characterized. These genes represent a diverse cluster of ontological functions including transcription factors, signaling molecules, and cell surface receptors. The expression levels of these genes were validated by Northern blot and/or quantitative RT-PCR analysis in both the JAK2 null cells and cells expressing a JAK2-dominant negative allele. As such, this work demonstrates for the first time that, in addition to being a key mediator of ligand-activated gene transcription, JAK2 can perhaps also be viewed as a critical mediator of basal level gene expression.


Subject(s)
Gene Expression Regulation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Line , Janus Kinase 2 , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...