Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Acad Med ; 98(11S): S14-S23, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37556802

ABSTRACT

PURPOSE: Basic science medical educators (BSME) play a vital role in the training of medical students, yet little is known about the factors that shape their professional identities. This multi-institutional qualitative study investigated factors that support and threaten the professional identity formation (PIF) of these medical educators. METHOD: A qualitative descriptive study was conducted with a purposive sample of 58 BSME from 7 allopathic medical schools in the U.S. In-depth semi-structured interviews of individual BSME were conducted between December 2020 and February 2021 to explore the facilitators and barriers shaping the PIF of BSME. Thematic analysis was conducted. RESULTS: Factors shaping PIF were grouped into 3 broad domains: personal, social, and structural. Interrelated themes described a combination of factors that pushed BSME into teaching (early or positive teaching experiences) and kept them there (satisfaction and rewards of teaching, communities of like-minded people), as well as factors that challenged their PIF (misunderstanding from medical students, clinical, and research faculty, lack of formal training programs, and lack of tenure-track educator positions). The structural environment was reported to be crucial for PIF and determined whether BSME felt that they belonged and were valued. CONCLUSIONS: This study shows that although most BSME derive a sense of fulfillment and meaning from their role as medical educators, they face considerable obstacles during their PIF. Structural change and support are needed to increase recognition, value, promotion, and belonging for BSME to improve the satisfaction and retention of this important group of faculty.


Subject(s)
Education, Medical, Undergraduate , Education, Medical , Humans , Social Identification , Faculty , Qualitative Research
2.
Pharmacol Res Perspect ; 9(3): e00762, 2021 05.
Article in English | MEDLINE | ID: mdl-33974344

ABSTRACT

Expectations for physicians are rapidly changing, as is the environment in which they will practice. In response, preclerkship medical education curricula are adapting to meet these demands, often by reducing the time for foundational sciences. This descriptive study compares preclerkship pharmacology education curricular practices from seven allopathic medical schools across the United States. We compare factors and practices that affect how pharmacology is integrated into the undergraduate medical education curriculum, including teaching techniques, resources, time allocated to pharmacology teaching, and assessment strategies. We use data from seven medical schools in the United States, along with results from a literature survey, to inform the strengths and weaknesses of various approaches and to raise important questions that can guide future research regarding integration of foundational sciences in medical school and health professions' curricula. In this comparative study, we found that there is significant heterogeneity in the number of hours dedicated to pharmacology in the preclerkship curriculum, whereas there was concordance in the use of active learning pedagogies for content delivery. Applying the ICAP (Interactive, Constructive, Active, Passive) Framework for cognitive engagement, our data showed that pharmacology was presented using more highly engaging pedagogies during sessions that are integrated with other foundational sciences. These findings can serve as a model that can be applied beyond pharmacology to other foundational sciences such as genetics, pathology, microbiology, biochemistry, etc.


Subject(s)
Curriculum , Education, Medical, Undergraduate , Pharmacology, Clinical/education , Schools, Medical , United States
3.
Pharmacol Res Perspect ; 9(3): e00773, 2021 05.
Article in English | MEDLINE | ID: mdl-33974347

ABSTRACT

A grounded knowledge of pharmacology is essential for healthcare providers to improve the quality of patients' lives, avoid medical errors, and circumvent potentially dangerous drug-drug interactions. One of the greatest tools to achieve this foundational knowledge of pharmacology is the dedicated pharmacology educators who teach in health sciences programs. Too often, the pharmacology educators responsible for teaching this material are left siloed at their own institutions with little room for dialog and collaboration. As scientists, we know that it is through dialog and collaboration that ideas grow, are refined, and improve. More collaborative work is needed to identify and describe best practices for pharmacology education in health sciences programs. While evidence-based, outcomes-focused studies are the optimum standard for this work, there is also a place for descriptive studies and innovative reports.


Subject(s)
Health Personnel/education , Interdisciplinary Placement , Pharmacology, Clinical/education , Curriculum , Humans
4.
Oncogene ; 39(19): 3791-3802, 2020 05.
Article in English | MEDLINE | ID: mdl-32203163

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDE) break down cyclic nucleotides such as cAMP and cGMP, reducing the signaling of these important intracellular second messengers. Several unique families of phosphodiesterases exist, and certain families are clinically important modulators of vasodilation. In the current work, we have summarized the body of literature that describes an emerging role for the PDE4 subfamily of phosphodiesterases in malignancy. We have systematically investigated PDE4A, PDE4B, PDE4C, and PDE4D isoforms and found evidence associating them with several cancer types including hematologic malignancies and lung cancers, among others. In this review, we compare the evidence examining the functional role of each PDE4 subtype across malignancies, looking for common signaling themes, signaling pathways, and establishing the case for PDE4 subtypes as a potential therapeutic target for cancer treatment.


Subject(s)
Hematologic Neoplasms/genetics , Lung Neoplasms/genetics , Protein Isoforms/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Hematologic Neoplasms/classification , Hematologic Neoplasms/pathology , Humans , Lung Neoplasms/classification , Lung Neoplasms/pathology , Protein Isoforms/classification , Signal Transduction/genetics
5.
Med Sci Educ ; 30(1): 111-115, 2020 Mar.
Article in English | MEDLINE | ID: mdl-34457648

ABSTRACT

Facebook is the social media platform used most by both medical educators and students, but there is scant literature investigating effective ways to use the platform across multiple institutions. This multi-institutional study compared student engagement between an official curricular Facebook page and a supplemental Facebook page. While a greater proportion of students were reached by the official course page, greater post engagement was achieved with the supplemental page. We attribute this to the abundance of question-type posts on the supplemental page and show that question-type posts achieve higher engagement with students than statement-type posts, regardless of page type.

6.
Curr Pharm Teach Learn ; 10(8): 1062-1069, 2018 08.
Article in English | MEDLINE | ID: mdl-30314542

ABSTRACT

BACKGROUND: Traditional interprofessional educational (IPE) exercises are those where learning exists "about, from, and with" trainees in two or more professions in order to prepare health sciences professionals to work on interprofessional teams. One emerging difficulty with IPE is the paucity of health profession students at single institutions, and the geographic and financial constraints of multi-institutional collaboration. INTERPROFESSIONAL EDUCATION ACTIVITY: To circumvent these barriers, we developed a multi-institution telehealth team-based learning (TBL) event between medical and pharmacy students on the topic of pharmacogenomics (PGx). Using a validated pre-post survey design, student attitudes and perceptions were measured before and after an educational intervention designed to simulate interprofessional telehealth collaboration. The survey results showed significant improvement across all areas of student attitudes toward interprofessional collaboration. Also, medical student PGx confidence increased substantially during the exercise even though the only PGx instruction they received was from pharmacy students. DISCUSSION: These data demonstrate that learning exists "about, from, and with" trainees in other professions, even if they do not physically train in the same location. Free tools are available to create virtual interactions between students on different campuses, and telehealth exercises using these tools are a valid way to conduct IPE across different campuses. The instructional experience does not need to be identical for all participants in the IPE event; rather, tailoring the educational experience to each group of students provides opportunities for inter-student teaching.


Subject(s)
Education, Distance/methods , Interprofessional Relations , Pharmacogenetics/education , Telemedicine/methods , Humans , Michigan , Problem-Based Learning/methods , Problem-Based Learning/trends , Students, Medical/psychology , Students, Medical/statistics & numerical data , Students, Pharmacy/psychology , Students, Pharmacy/statistics & numerical data , Teaching , User-Computer Interface
7.
J Invest Surg ; 28(5): 292-300, 2015.
Article in English | MEDLINE | ID: mdl-26374984

ABSTRACT

Liver ischemia reperfusion injury is mediated by a complex system of signaling cascades and inflammatory response resulting in organ damage. Selectins are a group of cell adhesion glycoproteins that play a key role in the initial immunological response. L-selectins, found on leukocytes, initiate the original adhesion and rolling phase of leukocyte extravasation upon liver sinusoidal endothelial cells (LSECs). P-selectins, found on platelets and tissue-specific endothelial cells, further increases leukocyte-endothelial adhesion and rolling. P-selectin-ligand binding also initiates intracellular signals that produce adhesion molecules to start firm adhesion and increase local chemokine production. L-selectin-ligand binding on the leukocytes increases adhesion molecule expression and chemokines, but also initiate changes in intracellular structural actin. E-selectin expression occurs with the presence of TNF-α and/or IL-1ß. E-selectin-ligand binding decreases leukocyte rolling velocity and increases adhesion molecules. Together, these glycoproteins transition the leukocyte response from original margination and rolling to firm adhesion and eventually migration.


Subject(s)
Liver Diseases/metabolism , Reperfusion Injury/metabolism , Selectins/metabolism , Animals , Humans , Liver Diseases/prevention & control , Reperfusion Injury/prevention & control
8.
Arch Toxicol ; 89(5): 651-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25566829

ABSTRACT

Ischemia/reperfusion (IR) injury occurs when oxygen is rapidly reintroduced into ischemic tissue, resulting in cell death and necrotic tissue damage. This is a major concern during liver transplantation procedures since there is an inevitable interruption and subsequent restoration of circulation. IR injury in liver tissue is initiated through reactive oxygen species (ROS), which are generated by hepatocytes during IR insult. Although these ROS are thought to play a protective roll since they are known to activate several pathways involved in the hypoxic response, they also trigger a localized sterile immune response that results in the recruitment of Kupffer cells and neutrophils to the site of IR insult. These immune cells generate larger quantities of ROS that trigger apoptosis and oncotic necrosis in liver tissue. In this review, we will summarize what is currently known about the response of liver tissue to IR insult at the molecular level.


Subject(s)
Ischemia/metabolism , Liver/blood supply , Reperfusion Injury/metabolism , Humans , Ischemia/etiology , Kupffer Cells/physiology , Liver/metabolism , Liver/pathology , Liver Transplantation/adverse effects , Necrosis , Reactive Oxygen Species/metabolism , Reperfusion Injury/etiology
9.
Clin Cancer Res ; 19(2): 380-92, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23213056

ABSTRACT

PURPOSE: Strategies to inhibit the EGF receptor (EGFR) using the tyrosine kinase inhibitor erlotinib have been associated with limited clinical efficacy in head and neck squamous cell carcinoma (HNSCC). Co-activation of alternative kinases may contribute to erlotinib resistance. EXPERIMENTAL DESIGN: We generated HNSCC cells expressing dominant-active c-Src (DA-Src) to determine the contribution of c-Src activation to erlotinib response. RESULTS: Expression of DA-Src conferred resistance to erlotinib in vitro and in vivo compared with vector-transfected control cells. Phospho-Met was strongly upregulated by DA-Src, and DA-Src cells did not produce hepatocyte growth factor (HGF). Knockdown of c-Met enhanced sensitivity to erlotinib in DA-Src cells in vitro, as did combining a c-Met or c-Src inhibitor with erlotinib. Inhibiting EGFR resulted in minimal reduction of phospho-Met in DA-Src cells, whereas complete phospho-Met inhibition was achieved by inhibiting c-Src. A c-Met inhibitor significantly sensitized DA-Src tumors to erlotinib in vivo, resulting in reduced Ki67 labeling and increased apoptosis. In parental cells, knockdown of endogenous c-Src enhanced sensitivity to erlotinib, whereas treatment with HGF to directly induce phospho-Met resulted in erlotinib resistance. The level of endogenous phospho-c-Src in HNSCC cell lines was also significantly correlated with erlotinib resistance. CONCLUSIONS: Ligand-independent activation of c-Met contributes specifically to erlotinib resistance, not cetuximab resistance, in HNSCC with activated c-Src, where c-Met activation is more dependent on c-Src than on EGFR, providing an alternate survival pathway. Addition of a c-Met or c-Src inhibitor to erlotinib may increase efficacy of EGFR inhibition in patients with activated c-Src.


Subject(s)
Head and Neck Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Quinazolines/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Enzyme Activation , Erlotinib Hydrochloride , Gene Expression , Gene Knockdown Techniques , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/genetics , Quinazolines/therapeutic use , Transcriptional Activation
10.
Cancer Biol Ther ; 13(10): 935-45, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22785204

ABSTRACT

The epidermal growth factor receptor (EGFR) is widely expressed in head and neck squamous cell carcinomas (HNSCC) and can activate many growth and survival pathways within tumor cells. Despite ubiquitous EGFR expression, therapies targeting the receptor are only modestly effective in the treatment of HNSCC. A consistent mechanism of resistance to EGFR targeting agents has not yet been identified in HNSCC likely due, in part, to the paucity of preclinical models. We assessed the in vitro and in vivo responses of a panel of 10 genotypically validated HNSCC cell lines to the EGFR inhibitors erlotinib and cetuximab to determine their validity as models of resistance to these agents. We defined a narrow range of response to erlotinib in HNSCC cells in vitro and found a positive correlation between EGFR protein expression and erlotinib response. We observed cross-sensitivity in one HNSCC cell line, 686LN, between erlotinib and cetuximab in vivo. We attempted to generate models of cetuximab resistance in HNSCC cell line-derived xenografts and heterotopic tumorgrafts generated directly from primary patient tumors. While all 10 HNSCC cell line xenografts tested were sensitive to cetuximab in vivo, heterotopic patient tumorgrafts varied in response to cetuximab indicating that these models may be more representative of clinical responses. These studies demonstrate the limitations of using HNSCC cell lines to reflect the heterogeneous clinical responses to erlotinib and cetuximab, and suggest that different approaches including heterotopic tumorgrafts may prove more valuable to elucidate mechanisms of clinical resistance to EGFR inhibitors in HNSCC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cetuximab , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Head and Neck Neoplasms/metabolism , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Squamous Cell Carcinoma of Head and Neck , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 17(18): 5935-44, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21791633

ABSTRACT

PURPOSE: Acquired resistance to cetuximab, a chimeric epidermal growth factor receptor (EGFR)-targeting monoclonal antibody, is a widespread problem in the treatment of solid tumors. The paucity of preclinical models has limited investigations to determine the mechanism of acquired therapeutic resistance, thereby limiting the development of effective treatments. The purpose of this study was to generate cetuximab-resistant tumors in vivo to characterize mechanisms of acquired resistance. EXPERIMENTAL DESIGN: We generated cetuximab-resistant clones from a cetuximab-sensitive bladder cancer cell line in vivo by exposing cetuximab-sensitive xenografts to increasing concentrations of cetuximab, followed by validation of the resistant phenotype in vivo and in vitro using invasion assays. A candidate-based approach was used to examine the role of HER2 on mediating cetuximab resistance both in vitro and in vivo. RESULTS: We generated a novel model of cetuximab resistance, and, for the first time in the context of EGFR-inhibitor resistance, we identified increased phosphorylation of a C-terminal fragment of HER2 (611-CTF) in cetuximab-resistant cells. Afatinib (BIBW-2992), an irreversible kinase inhibitor targeting EGFR and HER2, successfully inhibited growth of the cetuximab-resistant cells in vitro. When afatinib was combined with cetuximab in vivo, we observed an additive growth inhibitory effect in cetuximab-resistant xenografts. CONCLUSIONS: These data suggest that the use of dual EGFR-HER2 kinase inhibitors can enhance responses to cetuximab, perhaps in part due to downregulation of 611-CTF. This study conducted in a novel in vivo model provides a mechanistic rationale for ongoing phase I clinical trials using this combination treatment modality.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Silencing , Humans , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
12.
J Proteome Res ; 9(6): 3073-82, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20426488

ABSTRACT

Epidermal growth factor receptor (EGFR) is overexpressed in up to 90% of head and neck cancer (HNC), where increased expression levels of EGFR correlate with poor prognosis. To date, EGFR expression levels have not predicted the clinical response to the EGFR-targeting therapies. Elucidation of the molecular mechanisms underlying anti-EGFR-induced antitumor effects may shed some light on the mechanisms of HNC resistance to EGFR-targeting therapeutics and provide novel targets for improving the treatment of HNC. Here, we conducted a quantitative proteomics analysis to determine the molecular networks regulated by EGFR levels in HNC by specifically knocking-down EGFR and employing stable isotope labeling with amino acids in cell culture (SILAC). Following data normalization to minimize systematic errors and Western blotting validation, 12 proteins (e.g., p21, stratifin, and maspin) and 24 proteins (e.g., cdc2 and MTA2) were found to be significantly upregulated or downregulated by EGFR knockdown, respectively. Bioinformatic analysis revealed that these proteins were mainly involved in long-chain fatty acid biosynthesis and beta-oxidation, cholesterol biosynthesis, cell proliferation, DNA replication, and apoptosis. Cell cycle analysis confirmed that G(2)/M phase progression was significantly inhibited by EGFR knockdown, a hypothesis generated from network modeling. Further investigation of these molecular networks may not only enhance our understanding of the antitumor mechanisms of EGFR targeting but also improve patient selection and provide novel targets for better therapeutics.


Subject(s)
ErbB Receptors/genetics , ErbB Receptors/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Proteomics/methods , Blotting, Western , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cholesterol/metabolism , Fatty Acids/metabolism , Gene Knockdown Techniques , Head and Neck Neoplasms/enzymology , Humans , Isotope Labeling , Metabolic Networks and Pathways , Proteome/metabolism , RNA, Small Interfering/genetics , Reproducibility of Results , Signal Transduction
13.
Clin Cancer Res ; 15(11): 3740-50, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19470725

ABSTRACT

PURPOSE: We determined hepatocyte growth factor (HGF) and c-Met expression and signaling in human head and neck squamous cell carcinoma (HNSCC) cells and primary tissues and tested the ability of c-Met tyrosine kinase inhibitors (TKI) to block HGF-induced biological signaling. EXPERIMENTAL DESIGN: Expression and signaling were determined using immunoblotting, ELISA, and immunohistochemistry. Biological end points included wound healing, cell proliferation, and invasion. c-Met TKIs were tested for their ability to block HGF-induced signaling and biological effects in vitro and in xenografts established in nude mice. RESULTS: c-Met was expressed and functional in HNSCC cells. HGF was secreted by HNSCC tumor-derived fibroblasts, but not by HNSCC cells. Activation of c-Met promoted phosphorylation of AKT and mitogen-activated protein kinase as well as release of the inflammatory cytokine interleukin-8. Cell growth and wound healing were also stimulated by HGF. c-Met TKIs blocked HGF-induced signaling, interleukin-8 release, and wound healing. Enhanced invasion of HNSCC cells induced by the presence of tumor-derived fibroblasts was completely blocked with a HGF-neutralizing antibody. PF-2341066, a c-Met TKI, caused a 50% inhibition of HNSCC tumor growth in vivo with decreased proliferation and increased apoptosis within the tumors. In HNSCC tumor tissues, both HGF and c-Met protein were increased compared with expression in normal mucosa. CONCLUSIONS: These results show that HGF acts mainly as a paracrine factor in HNSCC cells, the HGF/c-Met pathway is frequently up-regulated and functional in HNSCC, and a clinically relevant c-Met TKI shows antitumor activity in vivo. Blocking the HGF/c-Met pathway may be clinically useful for the treatment of HNSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/physiopathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Crizotinib , Dose-Response Relationship, Drug , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/physiopathology , Hepatocyte Growth Factor/pharmacology , Humans , Immunohistochemistry , Indoles/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Paracrine Communication/physiology , Piperazines/pharmacology , Piperidines/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazoles , Pyridines/pharmacology , Signal Transduction/drug effects , Stress, Mechanical , Sulfonamides/pharmacology , Transplantation, Heterologous , Tumor Burden/drug effects
14.
Cancer Biol Ther ; 8(10): 907-16, 2009 May.
Article in English | MEDLINE | ID: mdl-19276661

ABSTRACT

The Cub and Sushi Multiple Domains-1 (CSMD1) is a tumor suppressor gene on 8p23.2, where allelic loss is both frequent and associated with poor prognosis in head and neck squamous cell carcinoma (HNSCC). To understand the extent of CSMD1 aberrations in vivo, we characterized 184 primary tumors from the head and neck, lung, breast and skin for gene copy number and analyzed expression in our HNSCCs and lung squamous cell carcinomas (SCCs). We detected loss of CSMD1 in a large proportion of HNSCCs (50%), lung (46%) and breast cancers (55%), and to a lesser extent in cutaneous SCCs (29%) and basal cell carcinomas (BCCs, 17%) using array-based comparative genomic hybridization (aCGH). Studying the region more closely with quantitative real-time PCR (qPCR), the loss of CSMD1 increased to 80% in HNSCCs and 93% in lung SCCs. CSMD1 expression was decreased in tumors compared to adjacent benign tissue (65%, 13/20) and was likely due to gene loss in 45% of cases (9/20). We also identified truncated transcripts lacking exons due to DNA copy number loss (30%, 5/17) or aberrant splicing (24%, 4/17). We show loss of CSMD1 in primary HNSCC tissues, and document for the first time that CSMD1 is lost in breast, lung and cutaneous SCCs. We also show that deletions of CSMD1 and aberrant splicing contribute to altered CSMD1 function in vivo.


Subject(s)
Breast Neoplasms/genetics , Head and Neck Neoplasms/genetics , Lung Neoplasms/genetics , Membrane Proteins/genetics , Skin Neoplasms/genetics , Breast Neoplasms/classification , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Chromosome Deletion , Comparative Genomic Hybridization , DNA, Neoplasm/analysis , Female , Gene Dosage , Gene Expression , Head and Neck Neoplasms/pathology , Humans , Loss of Heterozygosity , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Mouth Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Proteins
15.
J Cell Biochem ; 102(2): 311-9, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17661350

ABSTRACT

The epidermal growth factor receptor (EGFR) and signal transducers and activators of transcription (STATs) are commonly expressed and activated in many malignancies. EGFR is an upstream activator of several pathways involved in tumor progression, and STATs activate selected genes involved in oncogenesis. There are several different mechanisms by which STAT proteins can mediate intracellular EGFR signaling, including direct activation of STATs by EGFR binding and indirect activation of STATs through Src-mediated EGFR signaling. EGFR likely activates STAT in a manner distinctive from other mechanisms of STAT activation; STAT5 can be phosphorylated in an EGF-dependent manner at unique sites, conferring novel functions. Cumulative evidence suggests that targeting EGFR signaling pathways at several levels may demonstrate synergistic therapeutic effects compared with targeting the upstream receptor alone. Thus, methods to inhibit EGFR in conjunction with oncogenic STATs may represent a novel therapeutic strategy for cancers characterized by upregulation of EGFR signaling.


Subject(s)
ErbB Receptors/physiology , Neoplasms/metabolism , STAT Transcription Factors/physiology , Animals , ErbB Receptors/antagonists & inhibitors , Humans , Neoplasms/therapy , Phosphorylation , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
16.
Laryngoscope ; 116(5): 735-41, 2006 May.
Article in English | MEDLINE | ID: mdl-16652080

ABSTRACT

OBJECTIVES: Array-based comparative genomic hybridization (aCGH) was used to develop a genome-wide molecular profile of oral squamous cell carcinoma (OSCC). Copy number alterations (CNAs) were identified by chromosomal region, mapped to specific genes, and compared with several previously documented CNAs associated with head and neck squamous cell carcinoma (HNSCC). The status of 512 commonly altered cancer genes was assessed and evaluated as potential correlates of tumor behavior. METHODS: Tumor tissue DNA was isolated for aCGH from 21 prospectively collected fresh-frozen OSCC specimens. aCGH was performed at 0.9-Mb resolution to identify distinct regions of genomic alteration and their associated genes. Cancer genes commonly altered were then correlated with clinicopathologic tumor data. RESULTS: Genomic regions most frequently amplified (>35%) were located on 3q, 5p, 8q, 9q, and 20q, although regions most frequently deleted (>40%) involved chromosomes 3p, 8p, 13q, and 18q. Minimal regions of CNA identified, by aCGH narrowed larger, previously documented CNAs associated with HNSCC to significantly smaller regions, yielding shorter lists of candidate genes. Cancer-related genes altered in greater than 25% OSCC samples were identified (22 amplified, 17 deleted). Several genes associated with the Fanconi anemia DNA-damage response pathway were frequently altered, including BRCA1, BRCA2, FANCD2, and FANCG. Other cancer-related genes linked to hereditary cancer syndromes include VHL, MLH1, XPC, and RB1. CONCLUSIONS: Genome-wide aCGH can be used to detect and map CNAs in OSCC tissue specimens with high resolution. These data implicate several candidate genes and gene pathways in the tumorigenesis of sporadic OSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Genetic Predisposition to Disease , Genome , Mouth Neoplasms/genetics , Oncogenes/genetics , Carcinoma, Squamous Cell/pathology , Chromosome Aberrations/statistics & numerical data , DNA, Neoplasm/analysis , Female , Genes, Neoplasm , Humans , Male , Mouth Neoplasms/pathology , Nucleic Acid Hybridization , Prospective Studies , Sampling Studies , Sensitivity and Specificity , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...