Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael R. Emmert-Buck is active.

Publication


Featured researches published by Michael R. Emmert-Buck.


Science | 1996

Laser Capture Microdissection

Michael R. Emmert-Buck; Robert F. Bonner; Paul D. Smith; Rodrigo F. Chuaqui; Zhengping Zhuang; Seth R. Goldstein; Rhonda A. Weiss; Lance A. Liotta

Laser capture microdissection (LCM) under direct microscopic visualization permits rapid one-step procurement of selected human cell populations from a section of complex, heterogeneous tissue. In this technique, a transparent thermoplastic film (ethylene vinyl acetate polymer) is applied to the surface of the tissue section on a standard glass histopathology slide; a carbon dioxide laser pulse then specifically activates the film above the cells of interest. Strong focal adhesion allows selective procurement of the targeted cells. Multiple examples of LCM transfer and tissue analysis, including polymerase chain reaction amplification of DNA and RNA, and enzyme recovery from transferred tissue are demonstrated.


Oncogene | 2001

Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front.

Cloud P. Paweletz; Lu Charboneau; Verena E. Bichsel; Nicole L. Simone; Tina Chen; John W. Gillespie; Michael R. Emmert-Buck; Mark J. Roth; Emanuel F. Petricoin; Lance A. Liotta

Protein arrays are described for screening of molecular markers and pathway targets in patient matched human tissue during disease progression. In contrast to previous protein arrays that immobilize the probe, our reverse phase protein array immobilizes the whole repertoire of patient proteins that represent the state of individual tissue cell populations undergoing disease transitions. A high degree of sensitivity, precision and linearity was achieved, making it possible to quantify the phosphorylated status of signal proteins in human tissue cell subpopulations. Using this novel protein microarray we have longitudinally analysed the state of pro-survival checkpoint proteins at the microscopic transition stage from patient matched histologically normal prostate epithelium to prostate intraepithelial neoplasia (PIN) and then to invasive prostate cancer. Cancer progression was associated with increased phosphorylation of Akt (P<0.04), suppression of apoptosis pathways (P<0.03), as well as decreased phosphorylation of ERK (P<0.01). At the transition from histologically normal epithelium to PIN we observed a statistically significant surge in phosphorylated Akt (P<0.03) and a concomitant suppression of downstream apoptosis pathways which proceeds the transition into invasive carcinoma.


Trends in Genetics | 1998

Laser-capture microdissection: opening the microscopic frontier to molecular analysis.

Nicole L. Simone; Robert F. Bonner; John W. Gillespie; Michael R. Emmert-Buck; Lance A. Liotta

As the list of expressed human genes expands, a major scientific challenge is to understand the molecular events that drive normal tissue morphogenesis and the evolution of pathological lesions in actual tissue. Laser capture microdissection (LCM) has been developed to provide a reliable method to procure pure populations of cells from specific microscopic regions of tissue sections, in one step, under direct visualization. The cells of interest are transferred to a polymer film that is activated by laser pulses. The exact morphology of the procured cells (with intact DNA, RNA and proteins) is retained and held on the transfer film. With the advent of LCM, cDNA libraries can be developed from pure cells obtained directly from stained tissue, and microhybridization arrays of thousands of genes can now be used to examine gene expression in microdissected human tissue biopsies. The fluctuation of expressed genes or alterations in the cellular DNA that correlate with a particular disease stage can ultimately be compared within or between individual patients. Such a fingerprint of gene-expression patterns can provide crucial clues for etiology and might, ultimately, contribute to diagnostic decisions and therapies tailored to the individual patient. Molecules found to be associated with a defined pathological lesion might serve as imaging ot therapeutic targets.


Nature Genetics | 2002

Post-analysis follow-up and validation of microarray experiments

Rodrigo F. Chuaqui; Robert F. Bonner; Carolyn J.M. Best; John W. Gillespie; Michael J. Flaig; Stephen M. Hewitt; John Phillips; David B. Krizman; Michael A. Tangrea; Mamoun Ahram; W. Marston Linehan; Vladimir Knezevic; Michael R. Emmert-Buck

Measurement of gene-expression profiles using microarray technology is becoming increasingly popular among the biomedical research community. Although there has been great progress in this field, investigators are still confronted with a difficult question after completing their experiments: how to validate the large data sets that are generated? This review summarizes current approaches to verifying global expression results, discusses the caveats that must be considered, and describes some methods that are being developed to address outstanding problems.


American Journal of Pathology | 1999

Immuno-LCM: laser capture microdissection of immunostained frozen sections for mRNA analysis

Falko Fend; Michael R. Emmert-Buck; Rodrigo F. Chuaqui; Kristina A. Cole; Jeffrey Lee; Lance A. Liotta; Mark Raffeld

Microdissection of routinely stained or unstained frozen sections has been used successfully to obtain purified cell populations for the analysis of cell-specific gene expression patterns in primary tissues with a complex mixture of cell types. However, the precision and usefulness of microdissection is frequently limited by the difficulty to identify different cell types and structures by morphology alone. We therefore developed a rapid immunostaining procedure for frozen sections followed by laser capture microdissection (LCM) and RNA extraction, which allows targeted mRNA analysis of immunophenotypically defined cell populations. After fixation, frozen sections are immunostained under RNAse-free conditions using a rapid three-step streptavidin-biotin technique, dehydrated and immediately subjected to LCM. RNA is extracted from captured tissue, DNAse I treated, and reverse transcribed. Acetone-, methanol-, or ethanol/acetone-fixed sections give excellent immunostaining after 12 to 25 minutes total processing time. Specificity, precision, and speed of microdissection is markedly increased due to improved identification of desired (or undesired) cell types. The mRNA recovered from immunostained tissue is of high quality. Single-step PCR is able to amplify fragments of more than 600 bp from both housekeeping genes such as beta-actin as well as cell-specific messages such as CD4 or CD19, using cDNA derived from less than 500 immunostained, microdissected cells. Immuno-LCM allows specific mRNA analysis of cell populations isolated according to their immunophenotype or expression of function-related antigens and significantly expands our ability to investigate gene expression in heterogeneous tissues.


Molecular & Cellular Proteomics | 2002

2D Differential In-gel Electrophoresis for the Identification of Esophageal Scans Cell Cancer-specific Protein Markers

Ge Zhou; Hongmei Li; Dianne DeCamp; She Chen; Hongjun Shu; Yi Gong; Michael J. Flaig; John W. Gillespie; Nan Hu; Philip R. Taylor; Michael R. Emmert-Buck; Lance A. Liotta; Emanuel F. Petricoin; Yingming Zhao

The reproducibility of conventional two-dimensional (2D) gel electrophoresis can be improved using differential in-gel electrophoresis (DIGE), a new emerging technology for proteomic analysis. In DIGE, two pools of proteins are labeled with 1-(5-carboxypentyl)-1′-propylindocarbocyanine halide (Cy3) N-hydroxy-succinimidyl ester and 1-(5-carboxypentyl)-1′-methylindodi-carbocyanine halide (Cy5) N-hydroxysuccinimidyl ester fluorescent dyes, respectively. The labeled proteins are mixed and separated in the same 2D gel. 2D DIGE was applied to quantify the differences in protein expression between laser capture microdissection-procured esophageal carcinoma cells and normal epithelial cells and to define cancer-specific and normal-specific protein markers. Analysis of the 2D images from protein lysates of ∼ 250,000 cancer cells and normal cells identified 1038 protein spots in cancer cell lysates and 1088 protein spots in normal cell lysates. Of the detected proteins, 58 spots were up-regulated by >3-fold and 107 were down-regulated by >3-fold in cancer cells. In addition to previously identified down-regulated protein annexin I, tumor rejection antigen (gp96) was found up-regulated in esophageal squamous cell cancer. Global quantification of protein expression between laser capture-microdissected patient-matched cancer cells and normal cells using 2D DIGE in combination with mass spectrometry is a powerful tool for the molecular characterization of cancer progression and identification of cancer-specific protein markers.


Oncogene | 2000

Distinct pattern of expression of differentiation and growth-related genes in squamous cell carcinomas of the head and neck revealed by the use of laser capture microdissection and cDNA arrays

Chidchanok Leethanakul; Vyomesh Patel; John W. Gillespie; Michael Pallente; John F. Ensley; Sittichai Koontongkaew; Lance A. Liotta; Michael R. Emmert-Buck; J. Silvio Gutkind

Although risk factors for squamous cell carcinomas of the head and neck (HNSCC) are well recognized, very little is known about the molecular mechanisms responsible for this malignancy. Furthermore, the ability to investigate gene expression profiles at different stages of tumor progression is usually limited by the remarkable heterogeneity of these neoplastic lesions. Here, we show the successful use of laser capture microdissection (LCM) to procure specific cell populations. The 5000 cells from representative sets of HNSCC and their matching normal tissues are sufficient to extract RNA of high integrity for the synthesis of labeled amplified cDNA probes which can then be hybridized to these membranes arrayed with known human cancer-related cDNAs. Furthermore, when compared to normal tissues, we demonstrate a consistent decrease in expression of differentiation markers such as cytokeratins, and an increase in the expression of a number of signal transducing and cell cycle regulatory molecules, as well as growth and angiogenic factors and tissue degrading proteases. Unexpectedly, we also found that most HNSCC overexpress members of the wnt and notch growth and differentiation regulatory system, thus suggesting that the wnt and notch pathways may contribute in squamous cell carcinogenesis. This experimental approach may facilitate the identification candidate markers for the early detection of preneoplastic lesions, as well as novel targets for pharmacological intervention in this disease.


American Journal of Pathology | 2002

Evaluation of Non-Formalin Tissue Fixation for Molecular Profiling Studies

John W. Gillespie; Carolyn J.M. Best; Verena E. Bichsel; Kristina A. Cole; Susan F. Greenhut; Stephen M. Hewitt; Mamoun Ahram; Yvonne Gathright; Maria J. Merino; Robert L. Strausberg; Jonathan I. Epstein; Stanley R. Hamilton; Gallya Gannot; Galina V. Baibakova; Valerie S. Calvert; Michael J. Flaig; Rodrigo F. Chuaqui; Judi Herring; John Pfeifer; Emmanuel F. Petricoin; W. Marston Linehan; Paul H. Duray; G. Steven Bova; Michael R. Emmert-Buck

Using a general strategy for evaluating clinical tissue specimens, we found that 70% ethanol fixation and paraffin embedding is a useful method for molecular profiling studies. Human prostate and kidney were used as test tissues. The protein content of the samples was analyzed by one-dimensional gel electrophoresis, immunoblot, two-dimensional gel electrophoresis, and layered expression scanning. In each case, the fixed and embedded tissues produced results similar to that obtained from snap-frozen specimens, although the protein quantity was somewhat decreased. Recovery of mRNA was reduced in both quantity and quality in the ethanol-fixed samples, but was superior to that obtained from formalin-fixed samples and sufficient to perform reverse transcription polymerase chain reactions. Recovery of DNA from ethanol-fixed specimens was superior to formalin-fixed samples as determined by one-dimensional gel electrophoresis and polymerase chain reaction. In conclusion, specimens fixed in 70% ethanol and embedded in paraffin produce good histology and permit recovery of DNA, mRNA, and proteins sufficient for several downstream molecular analyses. Complete protocols and additional discussion of relevant issues are available on an accompanying website (http://cgap-mf.nih.gov/).


Electrophoresis | 2000

Proteomic analysis of laser capture microdissected human prostate cancer and in vitro prostate cell lines

David K. Ornstein; John W. Gillespie; Cloud P. Paweletz; Paul H. Duray; Judi Herring; Cathy D. Vocke; Suzanne L. Topalian; David G. Bostwick; W. Marston Linehan; Emanuel F. Petricoin; Michael R. Emmert-Buck

Specific populations of normal and malignant epithelium from three radical prostatectomy tissue specimes were procured by laser capture microdissection (LCM) and analyzed by two‐dimensional polyacrylamide gel electrophoresis (2‐D PAGE). Six proteins that were only seen in malignant cells and two proteins that were only seen in benign epithelium were reproducibly observed in two of two cases examined. Furthermore, these proteins were not observed in the 2‐D PAGE profiles from the patient‐matched microdissected stromal cell populations, but were seen in the protein profiles from the undissected whole cryostat sections. One of these proteins was determined to be prostate‐specific antigen (PSA) by Western blot analysis, and intriguingly the remaining protein candidates were found to be at least as abundant as the PSA protein. Comparison of 2‐D PAGE profiles of microdissected cell with matched in vitro cell lines from the same patient, and metastatic prostate cancer cell lines (LnCaP and PC3) showed striking differences between prostate cells in vivo and in vitro with less than 20% shared proteins. The data demonstrate that 2‐D PAGE analysis of LCM‐derived cells can reliably detect alterations in protein expression associated with prostate cancer, and that these differentially expressed proteins are produced in high enough levels which could allow for their clinical utility as new targets for therapeutic intervention, serum markers, and/or imaging markers.


Molecular Carcinogenesis | 2000

An approach to proteomic analysis of human tumors.

Michael R. Emmert-Buck; John W. Gillespie; Cloud P. Paweletz; David K. Ornstein; Venkatesha Basrur; Ettore Appella; Quan Hong Wang; Jing Huang; Nan Hu; Phil Taylor; Emanuel F. Petricoin

A strategy for proteomic analysis of microdissected cells derived from human tumor specimens is described and demonstrated by using esophageal cancer as an example. Normal squamous epithelium and corresponding tumor cells from two patients were procured by laser‐capture microdissection and studied by two‐dimensional polyacrylamide gel electrophoresis (2D‐PAGE). Fifty thousand cells resolved approximately 675 distinct proteins (or isoforms) with molecular weights ranging between 10 and 200 kDa and isoelectric points of pH 3–10. Comparison of the microdissected protein profiles showed a high degree of similarity between the matched normal‐tumor samples (98% identical). However, 17 proteins showed tumor‐specific alterations, including 10 that were uniquely present in the tumors and seven that were observed only in the normal epithelium. Two of the altered proteins were characterized by mass spectrometry and immunoblot analysis and were identified as cytokeratin 1 and annexin I. Acquisition of 2D‐PAGE protein profiles, visualization of disregulated proteins, and subsequent determination of the identity of selected proteins through high‐sensitivity MS‐MS microsequencing are possible from microdissected cell populations. These separation and analytical techniques are uniquely capable of detecting tumor‐specific alterations. Continued refinement of techniques and methodologies to determine the abundance and status of proteins in vivo holds great promise for future study of normal cells and associated neoplasms. Mol. Carcinog. 27:158–165, 2000. Published by Wiley‐Liss Inc.

Collaboration


Dive into the Michael R. Emmert-Buck's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rodrigo F. Chuaqui

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John W. Gillespie

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Jaime Rodriguez-Canales

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael A. Tangrea

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhengping Zhuang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

W. Marston Linehan

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Nan Hu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alisa M. Goldstein

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Heidi S. Erickson

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge