Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cancer Cell ; 42(5): 723-726, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38701793

ABSTRACT

Advances in biomedical research require a robust physician scientist workforce. Despite being equally successful at securing early career awards from the NIH as men, women MD-PhD physician scientists are less likely to serve as principal investigators on mid- and later careers awards. Here, we discuss the causes of gender disparities in academic medicine, the implications of losing highly trained women physician scientists, and the institutional and systemic changes needed to sustain this pool of talented investigators.


Subject(s)
Biomedical Research , Physicians, Women , Research Personnel , Humans , Female , Physicians, Women/statistics & numerical data , Male , Career Choice , United States , Sexism , Career Mobility , Physicians , Awards and Prizes
2.
Brain Pathol ; 33(3): e13118, 2023 05.
Article in English | MEDLINE | ID: mdl-36161399

ABSTRACT

Chronic inflammation is frequently invoked as a mechanism of neurodegeneration and yet inflammatory cell infiltrates are seldom seen in brains of these disorders. Different disciplines utilize different technologies and methodologies to describe what is immunologically defined as the innate immune response (IIR). We examined murine models of the human neurodegenerative disease Aicardi-Goutières Syndrome, where an IIR is initiated by aberrant RNA metabolism secondary to a mutation in adenosine deaminase acting on RNA gene (ADAR1). We previously showed that these mice demonstrated a deficit in RNA editing that lead to MDA-5 mediated RNA sensing pathway activation of the IIR with massive interferon stimulated gene transcription and translation. As early as 2 weeks of age, in situ hybridization demonstrated that different central nervous system (CNS) cell lineages expressed very high levels of distinct interferon stimulated genes (ISGs) in the absence of interferon and absence of immune cell infiltrates. We have expanded these studies to more completely describe the breadth of ISG expression systemically and in CNS using double label in situ hybridization. Within the CNS aberrant ISG expression was mostly limited to neurons, microglia, ependyma, choroid plexus, and endothelial cells with little expression in oligodendroglia and astrocytes except for STAT1. Wild type controls showed a similar pattern of ISG expression but only in aged mice and at levels minimally detectable by in situ hybridization. Despite months of elevated ISG expression in mutant mice, there was essentially no inflammatory infiltrate, no interferon production and minimal glial reaction. Histomorphological neurodegenerative pathology of ventricular dilatation and deep gray matter mineralization were evident in mutant mice 8-13 months of age but this did not show a spatial relationship to ISG expression. This IIR without immune cell infiltration leads to neurodegeneration through non-canonical pathways that may accentuate normal aging pathways.


Subject(s)
Endothelial Cells , Neurodegenerative Diseases , Humans , Animals , Mice , Endothelial Cells/metabolism , Disease Models, Animal , Neurodegenerative Diseases/metabolism , Brain/metabolism , Immunity, Innate , RNA/metabolism , Adenosine Deaminase/metabolism
3.
J Neuroinflammation ; 19(1): 285, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36457126

ABSTRACT

BACKGROUND: Aicardi-Goutières syndrome (AGS) is a severe neurodegenerative disease with clinical features of early-onset encephalopathy and progressive loss of intellectual abilities and motor control. Gene mutations in seven protein-coding genes have been found to be associated with AGS. However, the causative role of these mutations in the early-onset neuropathogenesis has not been demonstrated in animal models, and the mechanism of neurodegeneration of AGS remains ambiguous. METHODS: Via CRISPR/Cas-9 technology, we established a mutant mouse model in which a genetic mutation found in AGS patients at the ADAR1 coding gene (Adar) loci was introduced into the mouse genome. A mouse model carrying double gene mutations encoding ADAR1 and MDA-5 was prepared using a breeding strategy. Phenotype, gene expression, RNA sequencing, innate immune pathway activation, and pathologic studies including RNA in situ hybridization (ISH) and immunohistochemistry were used for characterization of the mouse models to determine potential disease mechanisms. RESULTS: We established a mouse model bearing a mutation in the catalytic domain of ADAR1, the D1113H mutation found in AGS patients. With this mouse model, we demonstrated a causative role of this mutation for the early-onset brain injuries in AGS and determined the signaling pathway underlying the neuropathogenesis. First, this mutation altered the RNA editing profile in neural transcripts and led to robust IFN-stimulated gene (ISG) expression in the brain. By ISH, the brains of mutant mice showed an unusual, multifocal increased expression of ISGs that was cell-type dependent. Early-onset astrocytosis and microgliosis and later stage calcification in the deep white matter areas were observed in the mutant mice. Brain ISG activation and neuroglial reaction were completely prevented in the Adar D1113H mutant mice by blocking RNA sensing through deletion of the cytosolic RNA receptor MDA-5. CONCLUSIONS: The Adar D1113H mutation in the ADAR1 catalytic domain results in early-onset and MDA5-dependent encephalopathy with IFN pathway activation in the mouse brain.


Subject(s)
Brain Injuries , Neurodegenerative Diseases , Animals , Mice , Catalytic Domain , Brain , Mutation/genetics , Disease Models, Animal , RNA , Adenosine Deaminase/genetics
4.
JCI Insight ; 7(6)2022 03 22.
Article in English | MEDLINE | ID: mdl-35315356

ABSTRACT

MD-PhD trainees constitute an important source of physician-scientists. Persistence on this challenging path is facilitated by success in garnering independent (R grant) support from the NIH. Published research tracks academic appointments and global R01 success for MD-PhD trainees but has not included information on future funding success of individual MD-PhD predoctoral grant holders. Here, we used data from the NIH RePORTER database to identify and track the funding trajectory of physician-scientists who received predoctoral grant support through the F30 mechanism, which is specific for dual-degree candidates. Male and female F30 awardees did not differ in their success in garnering K (postdoctoral training) grants, but, among F30 grant awardees, men were 2.6 times more likely than women to receive R funding. These results underscore the need for analysis of factors that contribute to the disproportionate loss of NIH-supported female physician-scientists between the predoctoral F30 and the independent R grant-supported stages.


Subject(s)
Biomedical Research , Physicians , Databases, Factual , Female , Humans , Male
5.
J Neuroinflammation ; 18(1): 169, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34332594

ABSTRACT

BACKGROUND: Aicardi-Goutières syndrome (AGS) is a severe infant or juvenile-onset autoimmune disease characterized by inflammatory encephalopathy with an elevated type 1 interferon-stimulated gene (ISG) expression signature in the brain. Mutations in seven different protein-coding genes, all linked to DNA/RNA metabolism or sensing, have been identified in AGS patients, but none of them has been demonstrated to activate the IFN pathway in the brain of an animal. The molecular mechanism of inflammatory encephalopathy in AGS has not been well defined. Adenosine Deaminase Acting on RNA 1 (ADAR1) is one of the AGS-associated genes. It carries out A-to-I RNA editing that converts adenosine to inosine at double-stranded RNA regions. Whether an AGS-associated mutation in ADAR1 activates the IFN pathway and causes autoimmune pathogenesis in the brain is yet to be determined. METHODS: Mutations in the ADAR1 gene found in AGS patients were introduced into the mouse genome via CRISPR/Cas9 technology. Molecular activities of the specific p.K999N mutation were investigated by measuring the RNA editing levels in brain mRNA substrates of ADAR1 through RNA sequencing analysis. IFN pathway activation in the brain was assessed by measuring ISG expression at the mRNA and protein level through real-time RT-PCR and Luminex assays, respectively. The locations in the brain and neural cell types that express ISGs were determined by RNA in situ hybridization (ISH). Potential AGS-related brain morphologic changes were assessed with immunohistological analysis. Von Kossa and Luxol Fast Blue staining was performed on brain tissue to assess calcification and myelin, respectively. RESULTS: Mice bearing the ADAR1 p.K999N were viable though smaller than wild type sibs. RNA sequencing analysis of neuron-specific RNA substrates revealed altered RNA editing activities of the mutant ADAR1 protein. Mutant mice exhibited dramatically elevated levels of multiple ISGs within the brain. RNA ISH of brain sections showed selective activation of ISG expression in neurons and microglia in a patchy pattern. ISG-15 mRNA was upregulated in ADAR1 mutant brain neurons whereas CXCL10 mRNA was elevated in adjacent astroglia. No calcification or gliosis was detected in the mutant brain. CONCLUSIONS: We demonstrated that an AGS-associated mutation in ADAR1, specifically the p.K999N mutation, activates the IFN pathway in the mouse brain. The ADAR1 p.K999N mutant mouse replicates aspects of the brain interferonopathy of AGS. Neurons and microglia express different ISGs. Basal ganglia calcification and leukodystrophy seen in AGS patients were not observed in K999N mutant mice, indicating that development of the full clinical phenotype may need an additional stimulus besides AGS mutations. This mutant mouse presents a robust tool for the investigation of AGS and neuroinflammatory diseases including the modeling of potential "second hits" that enable severe phenotypes of clinically variable diseases.


Subject(s)
Adenosine Deaminase/genetics , Autoimmune Diseases of the Nervous System/genetics , Brain/immunology , Immunity, Innate/genetics , Mutation , Nervous System Malformations/genetics , Animals , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/metabolism , Chemokines/metabolism , Cytokines/metabolism , Interferon Type I/immunology , Interferon Type I/metabolism , Mice , Nervous System Malformations/immunology , Nervous System Malformations/metabolism , RNA Editing
7.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33351788

ABSTRACT

Human lung adenocarcinoma (LUAD) in current or former smokers exhibits a high tumor mutational burden (TMB) and distinct mutational signatures. Syngeneic mouse models of clinically relevant smoking-related LUAD are lacking. We established and characterized a tobacco-associated, transplantable murine LUAD cell line, designated FVBW-17, from a LUAD induced by the tobacco carcinogen 4-(methylnitrosoamino)-1-(3-pyridyl)-1-butanone in the FVB/N mouse strain. Whole-exome sequencing of FVBW-17 cells identified tobacco-associated KrasG12D and Trp53 mutations and a similar mutation profile to that of classic alkylating agents with a TMB greater than 500. FVBW-17 cells transplanted subcutaneously, via tail vein, and orthotopically generated tumors that were histologically similar to human LUAD in FVB/N mice. FVBW-17 tumors expressed programmed death ligand 1 (PD-L1), were infiltrated with CD8+ T cells, and were responsive to anti-PD-L1 therapy. FVBW-17 cells were also engineered to express green fluorescent protein and luciferase to facilitate detection and quantification of tumor growth. Distant metastases to lung, spleen, liver, and kidney were observed from subcutaneously transplanted tumors. This potentially novel cell line is a robust representation of human smoking-related LUAD biology and provides a much needed preclinical model in which to test promising new agents and combinations, including immune-based therapies.


Subject(s)
Adenocarcinoma of Lung/chemically induced , B7-H1 Antigen/metabolism , Carcinogens/toxicity , Lung Neoplasms/chemically induced , Nitrosamines/toxicity , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Mice , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Smoke/adverse effects , Nicotiana/toxicity , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
8.
Acad Med ; 95(9): 1373-1381, 2020 09.
Article in English | MEDLINE | ID: mdl-32079926

ABSTRACT

The University of Pittsburgh School of Medicine Physician Scientist Training Program (PSTP) is a 5-year medical student training program designed to prepare the next generation of MD-only physician-scientists engaging in preclinical research. This article provides an overview of the program, including the novel longitudinal structure and competency goals, which facilitate success and persistence in a laboratory-based physician-scientist career. The authors present data on 81 medical students accepted to the program from academic year 2007-2008 through 2018-2019. Extrinsic outcomes, such as publications, grant funding, and residency matching, indicate that PSTP trainees have actively generated research deliverables. A majority of eligible PSTP trainees have earned Howard Hughes Medical Institute Medical Research Fellow funding. PSTP students have produced a mean of 1.6 first-authored publications (median, 1.0) and a mean of 5.1 total publications (median, 4.0) while in medical school and have authored 0.9 publications per year as residents/fellows, excluding internship. Nearly 60% of PSTP students (26/46) have matched to top-10 National Institutes of Health-funded residency programs in their specialty (based on Blue Ridge Institute rankings). PSTP alumni are twice as likely as their classmates to match into research-heavy departments and to publish first-authored papers. Results of a 2018 program evaluation survey indicate that intrinsic outcomes, such as confidence in research skills, significantly correlate with extrinsic outcomes. The program continues to evolve to maximize both scientific agency and career navigation skills in participants. This medical student PSTP model has potential to expand the pool of physician-scientist researchers in preclinical research beyond the capacity of dedicated MD-PhD and postgraduate training programs.


Subject(s)
Biomedical Research/education , Education, Graduate , Education, Medical , Internship and Residency , Pennsylvania , Physicians , Schools, Medical
9.
Int J Cancer ; 132(8): 1741-50, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-22987615

ABSTRACT

Patients with chronic myelogenous leukemia (CML) respond well to tyrosine kinase inhibitors (TKIs) of the Bcr-Abl oncoprotein. However, intolerance and resistance to these agents remains a challenge, and TKIs are unable to eradicate rare leukemia-initiating cells. Leukemia treatment would benefit from a better understanding of molecular signals that are necessary for the survival of leukemia-initiating cells but dispensable for normal hematopoietic stem cells. Leukemia-initiating cells in CML can arise from myeloid progenitor cells, a population that we have reported in normal hematopoiesis to depend on the RNA-editing enzyme adenosine deaminase acting on RNA-1 (ADAR1). We now report that Bcr-Abl transformed leukemic cells were ADAR1-dependent in a conditional ADAR1 knockout mouse model. ADAR1 deletion reversed leukocytosis and splenomegaly, and preferentially depleted primitive Lin-Sca+Kit+ (LSK) leukemic cells but not LSK cells lacking the leukemic oncoprotein. ADAR1 deletion ultimately normalized the peripheral white blood count, eliminating leukemic cells as assessed by PCR. These results uncover a novel requirement for ADAR1 in myeloid leukemic cells and indicate that ADAR1 may comprise a new molecular target for CML-directed therapeutics.


Subject(s)
Adenosine Deaminase/genetics , Gene Deletion , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Adenosine Deaminase Inhibitors/pharmacology , Animals , Base Sequence , DNA Primers , Flow Cytometry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology
10.
PLoS One ; 7(11): e48560, 2012.
Article in English | MEDLINE | ID: mdl-23185265

ABSTRACT

Heightened production of collagen and other matrix proteins underlies the fibrotic phenotype of systemic sclerosis (SSc). Roscovitine is an inhibitor of cyclin-dependent kinases that promote cell cycling (CDK1, 2), neuronal development (CDK5) and control transcription (CDK7,9). In an in vivo glomerulonephritis model, roscovitine treatment decreased mesangial cell proliferation and matrix proteins [1]. We investigated whether roscovitine could regulate fibrotic protein production directly rather than through cell cycling. Our investigations revealed that roscovitine coordinately inhibited the expression of collagen, fibronectin, and connective tissue growth factor (CTGF) in normal and SSc fibroblasts. This effect occurred on a transcriptional basis and did not result from roscovitine-mediated cell cycle inhibition. Roscovitine-mediated suppression of matrix proteins could not be reversed by the exogenous profibrotic cytokines TGF-ß or IL-6. To our knowledge, we are the first to report that roscovitine modulates matrix protein transcription. Roscovitine may thus be a viable treatment option for SSc and other fibrosing diseases.


Subject(s)
Fibroblasts/pathology , Purines/therapeutic use , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/pathology , Animals , Collagen/genetics , Collagen/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Fibroblasts/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis , Humans , Interleukin-6/biosynthesis , Mice , NIH 3T3 Cells , Phosphorylation/drug effects , Roscovitine , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , Up-Regulation/drug effects
11.
Breast Cancer Res ; 14(5): 213, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-23014660

ABSTRACT

Zoledronic acid (ZA) is an imidazole-containing bisphosphonate that has been extensively studied as an osteoclast inhibitor. ZA decreases bone turnover and has been effective in limiting osteolysis in metastatic cancers, including breast cancer. Recent clinical trials that demonstrated enhancement of disease-free survival by bisphosphonates have prompted interest in bisphosphonates as anti-cancer agents. ZA, for example, increased disease-free survival in postmenopausal and in premenopausal, hormone-suppressed breast cancer patients. Intriguingly, however, there was a lack of an anti-cancer effect of ZA in premenopausal women without ovarian suppression. These observations have prompted the conjecture that anti-cancer effects of ZA are limited to estrogen-poor environments. This review explores possible mechanisms compatible with differences in ZA activity in premenopausal women compared with postmenopausal (or hormone-suppressed) women.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Density Conservation Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Bone and Bones/drug effects , Bone and Bones/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Movement/drug effects , Diphosphonates/pharmacology , Drug Resistance, Neoplasm , Estrogens/metabolism , Female , Humans , Imidazoles/pharmacology , Immunity/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Signal Transduction , Tumor Microenvironment , Zoledronic Acid
14.
J Cancer Educ ; 25(3): 413-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20237886

ABSTRACT

There is a paucity of African-American Cancer researchers. To help address this, an educational collaboration was developed between a Comprehensive Cancer Center and a distant undergraduate biology department at a minority institution that sought to teach students introductory cancer biology while modeling research culture. A student-centered active learning curriculum was established that incorporated scientific poster presentations and simulated research exercises to foster learning of cancer biology. Students successfully mined primary literature for supportive data to test cancer-related hypotheses. Student feedback indicated that the poster project substantially enhanced depth of understanding of cancer biology and laid the groundwork for subsequent laboratory work. This inter-institutional collaboration modeled the research process while conveying facts and concepts about cancer.


Subject(s)
Biology/education , Biomedical Research/education , Interinstitutional Relations , Learning , Neoplasms/prevention & control , Documentation , Educational Measurement , Health Promotion , Humans , Posters as Topic , Students/psychology
15.
PLoS One ; 4(10): e7469, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19829692

ABSTRACT

The growth factor interleukin-3 (IL-3) promotes the survival and growth of multipotent hematopoietic progenitors and stimulates myelopoiesis. It has also been reported to oppose terminal granulopoiesis and to support leukemic cell growth through autocrine or paracrine mechanisms. The degree to which IL-3 acts at the posttranscriptional level is largely unknown. We have conducted global mRNA decay profiling and bioinformatic analyses in 32Dcl3 myeloblasts indicating that IL-3 caused immediate early stabilization of hundreds of transcripts in pathways relevant to myeloblast function. Stabilized transcripts were enriched for AU-Response elements (AREs), and an ARE-containing domain from the interleukin-6 (IL-6) 3'-UTR rendered a heterologous gene responsive to IL-3-mediated transcript stabilization. Many IL-3-stabilized transcripts had been associated with leukemic transformation. Deregulated Abl kinase shared with IL-3 the ability to delay turnover of transcripts involved in proliferation or differentiation blockade, relying, in part, on signaling through the Mek/Erk pathway. These findings support a model of IL-3 action through mRNA stability control and suggest that aberrant stabilization of an mRNA network linked to IL-3 contributes to leukemic cell growth.


Subject(s)
Gene Expression Regulation, Neoplastic , Granulocyte Precursor Cells/metabolism , Interleukin-3/metabolism , Proto-Oncogene Proteins c-abl/metabolism , RNA Stability/genetics , Amino Acid Motifs , Animals , Computational Biology/methods , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Kinase 1/metabolism , Mice , Oligonucleotide Array Sequence Analysis , RNA Processing, Post-Transcriptional , Response Elements
16.
Am J Pathol ; 175(2): 605-15, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19628764

ABSTRACT

We have previously shown that insulin-like growth factor (IGF) binding protein- 5 (IGFBP-5) is overexpressed in lung fibrosis and induces the production of extracellular matrix components, such as collagen and fibronectin, both in vitro and in vivo. The exact mechanism by which IGFBP-5 exerts these novel fibrotic effects is unknown. We thus examined the signaling cascades that mediate IGFBP-5-induced fibrosis. We demonstrate for the first time that IGFBP-5 induction of extracellular matrix occurs independently of IGF-I, and results from IGFBP-5 activation of MAPK signaling, which facilitates the translocation of IGFBP-5 to the nucleus. We examined the effects of IGFBP-5 on early growth response (Egr)-1, a transcription factor that is central to growth factor-mediated fibrosis. Egr-1 was up-regulated by IGFBP-5 in a MAPK-dependent manner and bound to nuclear IGFBP-5. In fibroblasts from Egr-1 knockout mice, induction of fibronectin by IGFBP-5 was abolished. Expression of Egr-1 in these cells rescued the extracellular matrix-promoting effects of IGFBP-5. Moreover, IGFBP-5 induced cell migration in an Egr-1-dependent manner. Notably, Egr-1 levels, similar to IGFBP-5, were increased in vivo in lung tissues and in vitro in primary fibroblasts of patients with pulmonary idiopathic fibrosis. Taken together, our findings suggest that IGFBP-5 induces a fibrotic phenotype via the activation of MAPK signaling and the induction of nuclear Egr-1 that interacts with IGFBP-5 and promotes fibrotic gene transcription.


Subject(s)
Early Growth Response Protein 1/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Pulmonary Fibrosis/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Movement , Cell Nucleus/metabolism , Early Growth Response Protein 1/genetics , Enzyme Activation , Fibronectins/biosynthesis , Humans , Insulin-Like Growth Factor Binding Protein 5/genetics , Mice , Mice, Knockout , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Transcription, Genetic
17.
J Biotechnol ; 143(2): 79-84, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19539675

ABSTRACT

Short hairpin RNA (shRNA)-expressing vectors have been shown stably to knock-down directed targets through RNA interference (RNAi). RNAi has emerged as a powerful tool for reverse genetic screens by assaying cellular phenotypes after exposure to libraries of pooled shRNAs directed against all or a subset of cellular mRNAs. Recently, noncoding RNAs have been recognized as major arbiters of cellular phenotypes. The scope and diversity of noncoding RNAs greatly enlarge the target pool for reversible genetic screens and underscore the desirability as screening tools of complex RNAi libraries, such as randomized RNAi libraries. We describe a novel approach to generate randomized shRNAs that takes advantage of a stable plasmid intermediate that arises in a FLP recombinase system. The ability of this system to generate randomized palindromes represents a new technology for shRNA generation including random shRNAs that cannot be produced synthetically. We describe this plasmid system and its use in generating randomized shRNAs from input 20-bp oligomers, and validate the functionality of a shRNA produced using this approach to knock-down estrogen receptor alpha expression.


Subject(s)
DNA Nucleotidyltransferases/genetics , Gene Knockdown Techniques/methods , Inverted Repeat Sequences , RNA, Small Interfering/genetics , Cloning, Molecular/methods , DNA Nucleotidyltransferases/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Gene Library , RNA, Small Interfering/metabolism , Reproducibility of Results
18.
Cell Cycle ; 5(24): 2899-902, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172866

ABSTRACT

Inactivation of the cyclin-dependent kinase (CDK) inhibitor p21(Waf1/Cip1) (CDKN1; hereafter p21) has previously been implicated in the induction of numerical centrosome alterations. It is unclear, however, whether p21 deficiency deregulates the centrosome duplication cycle itself or causes an accumulation of centrosomes due to cell division failure and/or polyploidization. Using a novel marker for maternal centrioles, Cep170, we show here that knock-down of p21 protein expression in murine myeloblasts can stimulate excessive centriole numbers in the presence of only one mature centriole. These results indicate that p21 deficiency can trigger a bona fide overduplication of centrioles and that aberrant centrosome numbers cannot solely be explained by polyploidization as suggested by previous studies. Our findings underscore that impaired p21 expression may function as a driving force for chromosomal instability and highlight the importance of markers for maternal centrioles such as Cep170 to elucidate the pathogenesis of numerical centriole aberrations in tumor cells.


Subject(s)
Centrioles/metabolism , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Animals , Granulocyte Precursor Cells/cytology , Mice , RNA, Small Interfering/metabolism
19.
Leuk Res ; 30(10): 1285-92, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16499966

ABSTRACT

Defining the molecular mechanisms that prevent myeloid progenitor cells from maturing is important because defects in maturation contribute to the development of myeloproliferative and myelodysplastic diseases. IL-3 is an important developmental factor for myeloid progenitor cells in vivo and is required to maintain the undifferentiated state in the 32Dcl3 cell line. The mechanisms employed by IL-3 to block differentiation, however, are not well understood. 32Dcl3 cells are myeloid progenitor cells of murine origin with high basal levels of p21waf1/cip1 (p21) expression. Our laboratory has previously reported that p21 levels decreased as CD34+-derived myeloid progenitor cells underwent terminal granulopoiesis in vitro. The effect of p21 upon the expression of genes associated with granulocytic differentiation has been unexplored, however. Since IL-3 maintains high levels of p21 in 32Dcl3 cells, we tested the hypothesis that p21 is an inhibitor of myeloid differentiation. Our findings demonstrate that siRNA knockdown of murine p21 is correlated with premature expression of the primary granule proteins myeloperoxidase and proteinase-3, proteins not abundant in cells maintained as myeloblasts by IL-3. Rescue with human p21 in these cells suppressed premature granule protein expression. p21 knockdown was also found to accelerate morphologic granulocytic differentiation in 32Dcl3 cells stimulated with G-CSF. Since high expression levels of p21 and overexpression of the IL-3 receptor have been correlated with poor outcomes in acute myeloid leukemias (AML), differentiation blockade by p21 may be one mechanism that contributes to AML pathogenesis.


Subject(s)
Cell Differentiation/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/cytology , Animals , Base Sequence , Blotting, Northern , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , DNA Primers , Gene Expression Regulation , Granulocytes/drug effects , Granulocytes/physiology , Interleukin-3/pharmacology , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Sequence Data , Plasmids , Polymerase Chain Reaction , Transcription, Genetic
20.
J Med Internet Res ; 6(3): e25, 2004 Sep 03.
Article in English | MEDLINE | ID: mdl-15471751

ABSTRACT

BACKGROUND: Adults with low literacy may encounter informational obstacles on the Internet when searching for health information, in part because most health Web sites require at least a high-school reading proficiency for optimal access. OBJECTIVE: The purpose of this study was to 1) determine how low-literacy adults independently access and evaluate health information on the Internet, 2) identify challenges and areas of proficiency in the Internet-searching skills of low-literacy adults. METHODS: Subjects (n=8) were enrolled in a reading assistance program at Bidwell Training Center in Pittsburgh, PA, and read at a 3rd to 8th grade level. Subjects conducted self-directed Internet searches for designated health topics while utilizing a think-aloud protocol. Subjects' keystrokes and comments were recorded using Camtasia Studio screen-capture software. The search terms used to find health information, the amount of time spent on each Web site, the number of Web sites accessed, the reading level of Web sites accessed, and the responses of subjects to questionnaires were assessed. RESULTS: Subjects collectively answered 8 out of 24 questions correctly. Seven out of 8 subjects selected "sponsored sites"-paid Web advertisements-over search engine-generated links when answering health questions. On average, subjects accessed health Web sites written at or above a 10th grade reading level. Standard methodologies used for measuring health literacy and for promoting subjects to verbalize responses to Web-site form and content had limited utility in this population. CONCLUSION: This study demonstrates that Web health information requires a reading level that prohibits optimal access by some low-literacy adults. These results highlight the low-literacy adult population as a potential audience for Web health information, and indicate some areas of difficulty that these individuals face when using the Internet and health Web sites to find information on specific health topics.


Subject(s)
Educational Status , Information Services , Internet/statistics & numerical data , Adult , Computer Literacy , Female , Humans , Male , Pennsylvania , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL
...