401
|
Janes KA, Reinhardt HC, Yaffe MB. Cytokine-induced signaling networks prioritize dynamic range over signal strength. Cell 2008; 135:343-54. [PMID: 18957207 DOI: 10.1016/j.cell.2008.08.034] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Revised: 06/12/2008] [Accepted: 08/16/2008] [Indexed: 12/28/2022]
Abstract
Signaling networks respond to diverse stimuli, but how the state of the signaling network is relayed to downstream cellular responses is unclear. We modeled how incremental activation of signaling molecules is transmitted to control apoptosis as a function of signal strength and dynamic range. A linear relationship between signal input and response output, with the dynamic range of signaling molecules uniformly distributed across activation states, most accurately predicted cellular responses. When nonlinearized signals with compressed dynamic range relay network activation to apoptosis, we observe catastrophic, stimulus-specific prediction failures. We develop a general computational technique, "model-breakpoint analysis," to analyze the mechanism of these failures, identifying new time- and stimulus-specific roles for Akt, ERK, and MK2 kinase activity in apoptosis, which were experimentally verified. Dynamic range is rarely measured in signal-transduction studies, but our experiments using model-breakpoint analysis suggest it may be a greater determinant of cell fate than measured signal strength.
Collapse
Affiliation(s)
- Kevin A Janes
- Koch Institute for Integrative Cancer Research, Center for Cell Decision Processes, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
402
|
Grafone T, Palmisano M, Nicci C, Martelli AM, Emanuela O, Storti S, Baccarani M, Martinelli G. Monitoring of FLT3 phosphorylation status and its response to drugs by flow cytometry in AML blast cells. Hematol Oncol 2008; 26:159-66. [PMID: 18383555 DOI: 10.1002/hon.854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
FLT3 mutation and overexpression in most acute myeloid leukaemia (AML) patients make this tyrosine kinase receptor an attractive therapeutic target. FLT3 kinase inhibitors are actually in clinical trials, thus it is critical to develop a reproducible and standardized method for screening of FLT3 activation and for monitoring its inhibition in response to drug in AML patients. We developed a flow cytometry method to analyse phosphorylated FLT3 (P-FLT3) in samples with <10(5) cells. The method was first validated in FLT3 wild-type (HL60/WT) and mutant (MV4-11/ITD(+)) as well as FLT3 negative (K562) cell lines. The method also proved to be reproducible in AML patient samples. Analysis was performed after exposure to drugs (CEP-701 and SU11657), in vitro and in vivo. In response to increasing drug concentrations, there was a linear reduction in P-FLT3. Intracellular flow cytometry analysis correlated with Western blot and XTT assays; flow cytometry data also correlated with FLT3 mutational status. The results highlight a rapid method to detect P-FLT3 protein at the single cell level by flow cytometry which enables an accurate assessment of FLT3 kinase activity in blast cells in response to novel tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Tiziana Grafone
- Institute of Hematology 'John Paul II' Centre for High Technology Research and Education in Biomedical Sciences, Catholic University, Campobasso, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
403
|
Cheong R, Wang CJ, Levchenko A. High content cell screening in a microfluidic device. Mol Cell Proteomics 2008; 8:433-42. [PMID: 18953019 DOI: 10.1074/mcp.m800291-mcp200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A comprehensive, systems level understanding of cell signaling networks requires methods to efficiently assay multiple signaling species, at the level of single cells, responding to a variety of stimulation protocols. Here we describe a microfluidic device that enables quantitative interrogation of signaling networks in thousands of individual cells using immunofluorescence-based readouts. The device is especially useful for measuring the signaling activity of kinases, transcription factors, and/or target genes in a high throughput, high content manner. We demonstrate how the device may be used to measure detailed time courses of signaling responses to one or more soluble stimuli and/or chemical inhibitors as well as responses to a complex temporal pattern of multiple stimuli. Furthermore we show how the throughput and resolution of the device may be exploited in investigating the differences, if any, of signaling at the level of a single cell versus at the level of the population. In particular, we show that NF-kappaB activity dynamics in individual cells are not asynchronous and instead resemble the dynamics of the population average in contrast to studies of cells overexpressing p65-EGFP.
Collapse
Affiliation(s)
- Raymond Cheong
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, Maryland 21218, USA
| | | | | |
Collapse
|
404
|
Kotecha N, Flores NJ, Irish JM, Simonds E, Sakai DS, Archambeault S, Diaz-Flores E, Coram M, Shannon KM, Nolan GP, Loh ML. Single-cell profiling identifies aberrant STAT5 activation in myeloid malignancies with specific clinical and biologic correlates. Cancer Cell 2008; 14:335-43. [PMID: 18835035 PMCID: PMC2647559 DOI: 10.1016/j.ccr.2008.08.014] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 07/24/2008] [Accepted: 08/29/2008] [Indexed: 12/31/2022]
Abstract
Progress in understanding the molecular pathogenesis of human myeloproliferative disorders (MPDs) has led to guidelines incorporating genetic assays with histopathology during diagnosis. Advances in flow cytometry have made it possible to simultaneously measure cell type and signaling abnormalities arising as a consequence of genetic pathologies. Using flow cytometry, we observed a specific evoked STAT5 signaling signature in a subset of samples from patients suspected of having juvenile myelomonocytic leukemia (JMML), an aggressive MPD with a challenging clinical presentation during active disease. This signature was a specific feature involving JAK-STAT signaling, suggesting a critical role of this pathway in the biological mechanism of this disorder and indicating potential targets for future therapies.
Collapse
MESH Headings
- Adult
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Cells, Cultured
- Child
- Disease Progression
- Flow Cytometry
- Gene Expression Regulation, Neoplastic
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Humans
- Janus Kinase 2/metabolism
- Leukemia, Myelomonocytic, Juvenile/genetics
- Leukemia, Myelomonocytic, Juvenile/metabolism
- Leukemia, Myelomonocytic, Juvenile/pathology
- Leukemia, Myelomonocytic, Juvenile/therapy
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/metabolism
- Myeloproliferative Disorders/pathology
- Myeloproliferative Disorders/therapy
- Neoplasm Staging
- Phosphorylation
- Recurrence
- STAT5 Transcription Factor/metabolism
- Signal Transduction/genetics
- Treatment Outcome
Collapse
Affiliation(s)
- Nikesh Kotecha
- Dept. of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305 USA
- Biomedical Informatics, Stanford University School of Medicine, Stanford, California 94305 USA
| | - Nikki J Flores
- Dept. of Pediatrics, University of California, San Francisco, San Francisco, California, 94143 USA
| | - Jonathan M Irish
- Dept. of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305 USA
- Dept. of Medicine, Stanford University School of Medicine, Stanford, California 94305 USA
| | - Erin Simonds
- Dept. of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305 USA
| | - Debbie S. Sakai
- Dept. of Pediatrics, University of California, San Francisco, San Francisco, California, 94143 USA
| | - Sophie Archambeault
- Dept. of Pediatrics, University of California, San Francisco, San Francisco, California, 94143 USA
| | - Ernesto Diaz-Flores
- Dept. of Pediatrics, University of California, San Francisco, San Francisco, California, 94143 USA
| | - Marc Coram
- BioStatistics, Stanford University School of Medicine, Stanford, California 94305 USA
| | - Kevin M Shannon
- Dept. of Pediatrics, University of California, San Francisco, San Francisco, California, 94143 USA
- UCSF Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, 94143 USA
| | - Garry P Nolan
- Dept. of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305 USA
- Correspondence should be addressed to, M.L.L (), G.P.N. ()
| | - Mignon L Loh
- Dept. of Pediatrics, University of California, San Francisco, San Francisco, California, 94143 USA
- UCSF Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, 94143 USA
- Correspondence should be addressed to, M.L.L (), G.P.N. ()
| |
Collapse
|
405
|
Abstract
Knowledge of the distinctive cellular and genetic traits of a cancer aids in diagnosis, prognosis, and potentially treatment. In this issue of Cancer Cell, Kotecha et al. (2008) demonstrate using a sophisticated flow cytometry approach that signal transduction responses to exogenous stimulation can inform diagnosis and pathobiology of myeloproliferative neoplasms.
Collapse
MESH Headings
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Child
- Disease Progression
- Flow Cytometry
- Gene Expression Regulation, Neoplastic
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Humans
- Janus Kinase 2/metabolism
- Leukemia, Myelomonocytic, Juvenile/genetics
- Leukemia, Myelomonocytic, Juvenile/metabolism
- Leukemia, Myelomonocytic, Juvenile/pathology
- Leukemia, Myelomonocytic, Juvenile/therapy
- Neoplasm Staging
- Phosphorylation
- STAT5 Transcription Factor/metabolism
- Signal Transduction/genetics
- Treatment Outcome
Collapse
Affiliation(s)
- Demetrios Kalaitzidis
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
406
|
STAT-3 and ERK 1/2 phosphorylation are critical for T-cell alloactivation and graft-versus-host disease. Blood 2008; 112:5254-8. [PMID: 18838616 DOI: 10.1182/blood-2008-03-147322] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a serious complication of allogeneic bone marrow transplantation, and donor T cells are indispensable for GVHD. Current therapies have limited efficacy, selectivity, and high toxicities. We used a novel flow cytometry technique for the analysis of intracellular phosphorylation events in single cells in murine BMT models to identify and validate novel GVHD drug targets.(1-7) This method circumvents the requirement for large numbers of purified cells, unlike western blots. We defined a signaling profile for alloactivated T cells in vivo and identified the phosphorylation of ERK1/2 and STAT-3 as important events during T-cell (allo)activation in GVHD. We establish that interference with STAT-3 phosphorylation can inhibit T-cell activation and proliferation in vitro and GVHD in vivo. This suggests that phospho-specific flow cytometry is useful for the identification of promising drug targets, and ERK1/2 and STAT-3 phosphorylation in alloactivated T cells may be important for GVHD.
Collapse
|
407
|
Aberrant GM-CSF signal transduction pathway in juvenile myelomonocytic leukemia assayed by flow cytometric intracellular STAT5 phosphorylation measurement. Leukemia 2008; 23:791-3. [DOI: 10.1038/leu.2008.265] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
408
|
Feinerman O, Veiga J, Dorfman JR, Germain RN, Altan-Bonnet G. Variability and robustness in T cell activation from regulated heterogeneity in protein levels. Science 2008; 321:1081-4. [PMID: 18719282 PMCID: PMC2673522 DOI: 10.1126/science.1158013] [Citation(s) in RCA: 278] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In T cells, the stochasticity of protein expression could contribute to the useful diversification of biological functions within a clonal population or interfere with accurate antigen discrimination. Combining computer modeling and single-cell measurements, we examined how endogenous variation in the expression levels of signaling proteins might affect antigen responsiveness during T cell activation. We found that the CD8 co-receptor fine-tunes activation thresholds, whereas the soluble hematopoietic phosphatase 1 (SHP-1) digitally regulates cell responsiveness. Stochastic variation in the expression of these proteins generates substantial diversity of activation within a clonal population of T cells, but co-regulation of CD8 and SHP-1 levels ultimately limits this very diversity. These findings reveal how eukaryotic cells can draw on regulated variation in gene expression to achieve phenotypic variability in a controlled manner.
Collapse
Affiliation(s)
- Ofer Feinerman
- ImmunoDynamics Group, Program in Computational Biology and Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue – Box 460, New York NY 10065 - USA
| | - Joël Veiga
- ImmunoDynamics Group, Program in Computational Biology and Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue – Box 460, New York NY 10065 - USA
| | - Jeffrey R. Dorfman
- ImmunoDynamics Group, Program in Computational Biology and Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue – Box 460, New York NY 10065 - USA
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immunology, Program in Systems Immunology and Infectious Disease Modeling (PSIIM), National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bldg. 10 Rm. 11N311, 10 Center Dr. MSC-1892, Bethesda, MD 20892-1892 – USA
| | - Grégoire Altan-Bonnet
- ImmunoDynamics Group, Program in Computational Biology and Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue – Box 460, New York NY 10065 - USA
| |
Collapse
|
409
|
Shachaf CM, Gentles AJ, Elchuri S, Sahoo D, Soen Y, Sharpe O, Perez OD, Chang M, Mitchel D, Robinson WH, Dill D, Nolan GP, Plevritis SK, Felsher DW. Genomic and proteomic analysis reveals a threshold level of MYC required for tumor maintenance. Cancer Res 2008; 68:5132-42. [PMID: 18593912 DOI: 10.1158/0008-5472.can-07-6192] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
MYC overexpression has been implicated in the pathogenesis of most types of human cancers. MYC is likely to contribute to tumorigenesis by its effects on global gene expression. Previously, we have shown that the loss of MYC overexpression is sufficient to reverse tumorigenesis. Here, we show that there is a precise threshold level of MYC expression required for maintaining the tumor phenotype, whereupon there is a switch from a gene expression program of proliferation to a state of proliferative arrest and apoptosis. Oligonucleotide microarray analysis and quantitative PCR were used to identify changes in expression in 3,921 genes, of which 2,348 were down-regulated and 1,573 were up-regulated. Critical changes in gene expression occurred at or near the MYC threshold, including genes implicated in the regulation of the G(1)-S and G(2)-M cell cycle checkpoints and death receptor/apoptosis signaling. Using two-dimensional protein analysis followed by mass spectrometry, phospho-flow fluorescence-activated cell sorting, and antibody arrays, we also identified changes at the protein level that contributed to MYC-dependent tumor regression. Proteins involved in mRNA translation decreased below threshold levels of MYC. Thus, at the MYC threshold, there is a loss of its ability to maintain tumorigenesis, with associated shifts in gene and protein expression that reestablish cell cycle checkpoints, halt protein translation, and promote apoptosis.
Collapse
Affiliation(s)
- Catherine M Shachaf
- Department of Medicine and Pathology, Division of Medical Oncology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
410
|
LaPan P, Zhang J, Pan J, Hill A, Haney SA. Single cell cytometry of protein function in RNAi treated cells and in native populations. BMC Cell Biol 2008; 9:43. [PMID: 18673568 PMCID: PMC2529295 DOI: 10.1186/1471-2121-9-43] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Accepted: 08/01/2008] [Indexed: 01/10/2023] Open
Abstract
Background High Content Screening has been shown to improve results of RNAi and other perturbations, however significant intra-sample heterogeneity is common and can complicate some analyses. Single cell cytometry can extract important information from subpopulations within these samples. Such approaches are important for immune cells analyzed by flow cytometry, but have not been broadly available for adherent cells that are critical to the study of solid-tumor cancers and other disease models. Results We have directly quantitated the effect of resolving RNAi treatments at the single cell level in experimental systems for both exogenous and endogenous targets. Analyzing the effect of an siRNA that targets GFP at the single cell level permits a stronger measure of the absolute function of the siRNA by gating to eliminate background levels of GFP intensities. Extending these methods to endogenous proteins, we have shown that well-level results of the knockdown of PTEN results in an increase in phospho-S6 levels, but at the single cell level, the correlation reveals the role of other inputs into the pathway. In a third example, reduction of STAT3 levels by siRNA causes an accumulation of cells in the G1 phase of the cell cycle, but does not induce apoptosis or necrosis when compared to control cells that express the same levels of STAT3. In a final example, the effect of reduced p53 levels on increased adriamycin sensitivity for colon carcinoma cells was demonstrated at the whole-well level using siRNA knockdown and in control and untreated cells at the single cell level. Conclusion We find that single cell analysis methods are generally applicable to a wide range of experiments in adherent cells using technology that is becoming increasingly available to most laboratories. It is well-suited to emerging models of signaling dysfunction, such as oncogene addition and oncogenic shock. Single cell cytometry can demonstrate effects on cell function for protein levels that differ by as little as 20%. Biological differences that result from changes in protein level or pathway activation state can be modulated directly by RNAi treatment or extracted from the natural variability intrinsic to cells grown under normal culture conditions.
Collapse
Affiliation(s)
- Peter LaPan
- Department of Biological Technologies, Oncology Research, Wyeth Research, 87 Cambridge Park Drive, Cambridge, MA 02140, USA.
| | | | | | | | | |
Collapse
|
411
|
White FM. Quantitative phosphoproteomic analysis of signaling network dynamics. Curr Opin Biotechnol 2008; 19:404-9. [PMID: 18619541 DOI: 10.1016/j.copbio.2008.06.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2008] [Accepted: 06/15/2008] [Indexed: 01/19/2023]
Abstract
Protein phosphorylation mediated cellular signaling is a highly regulated, dynamic process that controls many aspects of cellular biology. Over the past few years many methods have been developed to quantify temporal dynamics of protein phosphorylation, including mass spectrometry, which can be applied in both an unbiased, discovery mode and in a targeted mode to monitor specific phosphorylation sites. Other methods, such as kinase activity assays and antibody microarrays, have been applied to quantify central nodes in the signaling network, yielding intriguing biological insights. This review provides a concise overview of the latest advances in the quantitative analysis of signaling dynamics including a brief commentary on the future of the field.
Collapse
Affiliation(s)
- Forest M White
- Department of Biological Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Building 56-787a, 77 Massachusetts Avenue, Cambridge, MA 02139, United States.
| |
Collapse
|
412
|
Abstract
Diverse cellular processes are regulated by tyrosyl phosphorylation, which is controlled by protein-tyrosine kinases (PTKs) and protein-tyrosine phosphatases (PTPs). De-regulated tyrosyl phosphorylation, evoked by gain-of-function mutations and/or over-expression of PTKs, contributes to the pathogenesis of many cancers and other human diseases. PTPs, because they oppose the action of PTKs, had been considered to be prime suspects for potential tumor suppressor genes. Surprisingly, few, if any, tumor suppressor PTPs have been identified. However, the Src homology-2 domain-containing phosphatase Shp2 (encoded by PTPN11) is a bona fide proto-oncogene. Germline mutations in PTPN11 cause Noonan and LEOPARD syndromes, whereas somatic PTPN11 mutations occur in several types of hematologic malignancies, most notably juvenile myelomonocytic leukemia and, more rarely, in solid tumors. Shp2 also is an essential component in several other oncogene signaling pathways. Elucidation of the events underlying Shp2-evoked transformation may provide new insights into oncogenic mechanisms and novel targets for anti-cancer therapy.
Collapse
|
413
|
Hamadani M, Awan FT, Copelan EA. Hematopoietic stem cell transplantation in adults with acute myeloid leukemia. Biol Blood Marrow Transplant 2008; 14:556-67. [PMID: 18410898 DOI: 10.1016/j.bbmt.2008.02.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Accepted: 02/27/2008] [Indexed: 11/26/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is an integral part of the treatment of many patients with acute myeloid leukemia (AML). Despite extensive study, the appropriate role and timing of allogeneic and autologous transplantation in AML are poorly defined. This review critically analyzes the extensive literature, focusing on the recent advances, and provides practical recommendations for the use of HSCT in AML.
Collapse
Affiliation(s)
- Mehdi Hamadani
- Division of Hematology & Oncology, Arthur G. James Cancer Hospital, Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
414
|
Schulz KR, Danna EA, Krutzik PO, Nolan GP. Single-cell phospho-protein analysis by flow cytometry. ACTA ACUST UNITED AC 2008; Chapter 8:8.17.1-8.17.20. [PMID: 18432997 DOI: 10.1002/0471142735.im0817s78] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This protocol describes methods for monitoring intracellular phosphorylation-dependent signaling events on a single-cell basis. This approach measures cell signaling by treating cells with exogenous stimuli, fixing cells with formaldehyde, permeabilizing with methanol, and then staining with phospho-specific antibodies. Thus, cell signaling states can be determined as a measure of how cells interact with their environment. This method has applications in clinical research as well as mechanistic studies of basic biology. In clinical research, diagnostic or drug efficacy information can be retrieved by discovering how a disease affects the ability of cells to respond to growth factors. Basic scientists can use this technique to analyze signaling events in cell lines and human or murine primary cells, including rare populations, like B1 cells or stem cells. This technique has broad applications to take standard biochemical analysis into primary cells to garner valuable information about signaling events in physiologic settings.
Collapse
|
415
|
Wen J, Yang X, Wang K, Tan W, Zuo X, Zhang H. Telomerase catalyzed fluorescent probes for sensitive protein profiling based on one-dimensional microfluidic beads array. Biosens Bioelectron 2008; 23:1788-92. [DOI: 10.1016/j.bios.2008.02.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 02/20/2008] [Accepted: 02/20/2008] [Indexed: 11/15/2022]
|
416
|
Schweneker M, Favre D, Martin JN, Deeks SG, McCune JM. HIV-induced changes in T cell signaling pathways. THE JOURNAL OF IMMUNOLOGY 2008; 180:6490-500. [PMID: 18453567 DOI: 10.4049/jimmunol.180.10.6490] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection with HIV usually results in chronic activation of the immune system, with profound quantitative and qualitative changes in the T cell compartment. To better understand the mechanistic basis for T cell dysfunction and to discern whether such mechanisms are reversed after effective antiviral treatment, we analyzed changes in signaling pathways of human CD4(+) and CD8(+) T cells from 57 HIV-infected subjects in varying stages of disease progression and treatment, including long-term nonprogressors, progressors, and chronically infected subjects provided effective antiretroviral therapy (responders). A previously described PhosFlow method was adapted and optimized so that protein phosphorylation could be visualized in phenotypically defined subpopulations of CD4(+) and CD8(+) T cells (naive, memory, and effector) by flow cytometry. T cell signaling induced by TCR cross-linking, IL-2, or PMA/ionomycin was found to be blunted within all T cell subpopulations in those with progressive HIV disease compared with long-term nonprogressors and responders. Although alterations in cellular signaling correlated with levels of basal phosphorylation, viral load, and/or expression of programmed death-1, it was the level of basal phosphorylation that appeared to be the factor most dominantly associated with impaired signaling. Notably, provision of effective antiretroviral therapy was associated with a normalization of both basal phosphorylation levels and T cell signaling. These data, in aggregate, suggest that generalized dysfunction of the T cell compartment during progressive HIV disease may be in part dependent upon an increased basal level of phosphorylation, which itself may be due to the heightened state of immune activation found in advanced disease.
Collapse
Affiliation(s)
- Marc Schweneker
- Division of Experimental Medicine, University of California, San Francisco, CA 94110, USA
| | | | | | | | | |
Collapse
|
417
|
van Baal JWPM, Krishnadath KK. High throughput techniques for characterizing the expression profile of Barrett's esophagus. Dis Esophagus 2008; 21:634-40. [PMID: 18564162 DOI: 10.1111/j.1442-2050.2008.00853.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Barrett's esophagus (BE) is the metaplastic change of the normal lined squamous epithelium of the distal esophagus to a columnar type of epithelium as a result of chronic long-standing gastroesophageal reflux disease. Patients with BE have a significantly increased risk of developing an esophageal adenocarcinoma, with an estimated annual incidence varying from 0.4 to 1.8%. Over the last 3 decades, the incidence of BE and its associated adenocarcinoma has increased in Western countries at a rate that exceeds that of any other malignancy. Despite all the research performed on BE, there is still an inadequate understanding of the biological basis of this mucosal transformation. With the upcoming modern high throughput technologies, important progression has been made in unraveling the expression profiles and gaining more insight in the biology of BE and esophageal adenocarcinoma. Several studies reported genome, transcriptome, proteome, and kinome investigations using high throughput techniques. These studies were conducted to find biomarkers that can be used to detect BE patients with increased risk for malignant progression or to obtain more insight in the mechanism underlying BE development. In the following review, we first discuss the different techniques that are currently available and summarize findings in this field, including several recent publications of our group.
Collapse
Affiliation(s)
- J W P M van Baal
- Laboratory of Experimental Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| | | |
Collapse
|
418
|
Abstract
Malignant tumor impairs human's health severely. Although progresses have been made in tumor's etiology, clinical management of tumor is still confronted by challenges. The standard treatments against tumor, including surgery, chemotherapy and radiotherapy, are not effective for all patients. The clinicians often feel confused while selecting appropriate therapeutic pattern and judging patients' prognosis. The clinical outcome of target-protein-based cancer therapy revealed that personalized molecular signature determined individualized curative effect. Effective management of tumor needs patient-tailored design. The so-called "omic" researches accelerate the studies in specific molecule phenotype of tumor cells, and the personalized therapy against such individualized malignant phenotype will represent the future direction in tumor treatment.
Collapse
|
419
|
Subkhankulova T, Gilchrist MJ, Livesey FJ. Modelling and measuring single cell RNA expression levels find considerable transcriptional differences among phenotypically identical cells. BMC Genomics 2008; 9:268. [PMID: 18522723 PMCID: PMC2429916 DOI: 10.1186/1471-2164-9-268] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 06/03/2008] [Indexed: 11/10/2022] Open
Abstract
Background Phenotypically identical cells demonstrate predictable, robust behaviours. However, there is uncertainty as to whether phenotypically identical cells are equally similar at the underlying transcriptional level or if cellular systems are inherently noisy. To answer this question, it is essential to distinguish between technical noise and true variation in transcript levels. A critical issue is the contribution of sampling effects, introduced by the requirement to globally amplify the single cell mRNA population, to observed measurements of relative transcript abundance. Results We used single cell microarray data to develop simple mathematical models, ran Monte Carlo simulations of the impact of technical and sampling effects on single cell expression data, and compared these with experimental microarray data generated from single embryonic neural stem cells in vivo. We show that the actual distribution of measured gene expression ratios for pairs of neural stem cells is much broader than that predicted from our sampling effect model. Conclusion Our results confirm that significant differences in gene expression levels exist between phenotypically identical cells in vivo, and that these differences exceed any noise contribution from global mRNA amplification.
Collapse
Affiliation(s)
- Tatiana Subkhankulova
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1 QN, UK.
| | | | | |
Collapse
|
420
|
Shao DD, Suresh R, Vakil V, Gomer RH, Pilling D. Pivotal Advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation. J Leukoc Biol 2008; 83:1323-33. [PMID: 18332234 PMCID: PMC2659591 DOI: 10.1189/jlb.1107782] [Citation(s) in RCA: 214] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD14+ peripheral blood monocytes can differentiate into fibroblast-like cells called fibrocytes, which are associated with and are at least partially responsible for wound healing and fibrosis in multiple organ systems. Signals regulating fibrocyte differentiation are poorly understood. In this study, we find that when added to human PBMCs cultured in serum-free medium, the profibrotic cytokines IL-4 and IL-13 promote fibrocyte differentiation without inducing fibrocyte or fibrocyte precursor proliferation. We also find that the potent, antifibrotic cytokines IFN-gamma and IL-12 inhibit fibrocyte differentiation. In our culture system, IL-1beta, IL-3, IL-6, IL-7, IL-16, GM-CSF, M-CSF, fetal liver tyrosine kinase 3, insulin growth factor 1, vascular endothelial growth factor, and TNF-alpha had no significant effect on fibrocyte differentiation. IL-4, IL-13, and IFN-gamma act directly on monocytes to regulate fibrocyte differentiation, and IL-12 acts indirectly, possibly through CD16-positive NK cells. We previously identified the plasma protein serum amyloid P (SAP) as a potent inhibitor of fibrocyte differentiation. When added together, the fibrocyte-inhibitory activity of SAP dominates the profibrocyte activities of IL-4 and IL-13. The profibrocyte activities of IL-4 and IL-13 and the fibrocyte-inhibitory activities of IFN-gamma and IL-12 counteract each other in a concentration-dependent manner. These results indicate that the complex mix of cytokines and plasma proteins present in inflammatory lesions, wounds, and fibrosis will influence fibrocyte differentiation.
Collapse
Affiliation(s)
- Diane D. Shao
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
| | - Rahul Suresh
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
| | - Varsha Vakil
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
| | - Richard H. Gomer
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
| | - Darrell Pilling
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
| |
Collapse
|
421
|
Siebert JC, Inokuma M, Waid DM, Pennock ND, Vaitaitis GM, Disis ML, Dunne JF, Wagner DH, Maecker HT. An analytical workflow for investigating cytokine profiles. Cytometry A 2008; 73:289-98. [PMID: 18163472 DOI: 10.1002/cyto.a.20509] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Understanding cytokine profiles of disease states has provided researchers with great insight into immunologic signaling associated with disease onset and progression, affording opportunities for advancement in diagnostics and therapeutic intervention. Multiparameter flow cytometric assays support identification of specific cytokine secreting subpopulations. Bead-based assays provide simultaneous measurement for the production of ever-growing numbers of cytokines. These technologies demand appropriate analytical techniques to extract relevant information efficiently. We illustrate the power of an analytical workflow to reveal significant alterations in T-cell cytokine expression patterns in type 1 diabetes (T1D) and breast cancer. This workflow consists of population-level analysis, followed by donor-level analysis, data transformation such as stratification or normalization, and a return to population-level analysis. In the T1D study, T-cell cytokine production was measured with a cytokine bead array. In the breast cancer study, intracellular cytokine staining measured T cell responses to stimulation with a variety of antigens. Summary statistics from each study were loaded into a relational database, together with associated experimental metadata and clinical parameters. Visual and statistical results were generated with custom Java software. In the T1D study, donor-level analysis led to the stratification of donors based on unstimulated cytokine expression. The resulting cohorts showed statistically significant differences in poststimulation production of IL-10, IL-1 beta, IL-8, and TNF beta. In the breast cancer study, the differing magnitude of cytokine responses required data normalization to support statistical comparisons. Once normalized, data showed a statistically significant decrease in the expression of IFN gamma on CD4+ and CD8+ T cells when stimulated with tumor-associated antigens (TAAs) when compared with an infectious disease antigen stimulus, and a statistically significant increase in expression of IL-2 on CD8+ T cells. In conclusion, the analytical workflow described herein yielded statistically supported and biologically relevant findings that were otherwise unapparent.
Collapse
Affiliation(s)
- Janet C Siebert
- CytoAnalytics, Analytical Services, Denver, Colorado 80209, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
422
|
Liu F, Kunter G, Krem MM, Eades WC, Cain JA, Tomasson MH, Hennighausen L, Link DC. Csf3r mutations in mice confer a strong clonal HSC advantage via activation of Stat5. J Clin Invest 2008; 118:946-55. [PMID: 18292815 DOI: 10.1172/jci32704] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Accepted: 12/19/2007] [Indexed: 12/11/2022] Open
Abstract
A fundamental property of leukemic stem cells is clonal dominance of the bone marrow microenvironment. Truncation mutations of CSF3R, which encodes the G-CSF receptor (G-CSFR), are implicated in leukemic progression in patients with severe congenital neutropenia. Here we show that expression of a truncated mutant Csf3r in mice confers a strong clonal advantage at the HSC level that is dependent upon exogenous G-CSF. G-CSF-induced proliferation, phosphorylation of Stat5, and transcription of Stat5 target genes were increased in HSCs isolated from mice expressing the mutant Csf3r. Conversely, the proliferative advantage conferred by the mutant Csf3r was abrogated in myeloid progenitors lacking both Stat5A and Stat5B, and HSC function was reduced in mice expressing a truncated mutant Csf3r engineered to have impaired Stat5 activation. These data indicate that in mice, inappropriate Stat5 activation plays a key role in establishing clonal dominance by stem cells expressing mutant Csf3r.
Collapse
Affiliation(s)
- Fulu Liu
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
423
|
Hedley DW, Chow S, Goolsby C, Shankey TV. Pharmacodynamic monitoring of molecular-targeted agents in the peripheral blood of leukemia patients using flow cytometry. Toxicol Pathol 2008; 36:133-9. [PMID: 18337231 DOI: 10.1177/0192623307310952] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The introduction of specific, molecular-targeted drugs is radically changing cancer treatment. Pharmacodynamics, which measures drug effects on the host, is key during early-phase clinical trials of novel agents to determine the relations between drug dose and target inhibition as well as measure the downstream effects of target inhibition on the cancer. In this article, we describe the application of flow cytometry to the pharmacodynamic monitoring of molecular-targeted agents in leukemia patients. The methods are based on current clinical flow-cytometry applications, with the addition of phosphospecific antibodies to measure the activation states of intracellular signaling elements and the introduction of techniques that maintain drug-target equilibrium during sample preparation. Using this approach, we successfully showed dose-dependent inhibition of c-Kit during a phase I clinical trial treating acute leukemia patients with the novel agent sorafenib. Further refinements identify considerable interpatient variation in signaling activity within leukemic blast populations, suggesting that an individualized approach to treatment based on flow cytometric monitoring might be advantageous. Improvements in sample turnaround offer the potential to introduce real-time pharmacodynamic monitoring during early-phase clinical trials.
Collapse
Affiliation(s)
- David W Hedley
- Division of Applied Molecular Oncology, Ontario Cancer Institute, University of Toronto, Ontario, Canada.
| | | | | | | |
Collapse
|
424
|
Zamir E, Geiger B, Kam Z. Quantitative multicolor compositional imaging resolves molecular domains in cell-matrix adhesions. PLoS One 2008; 3:e1901. [PMID: 18382676 PMCID: PMC2270910 DOI: 10.1371/journal.pone.0001901] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2007] [Accepted: 02/20/2008] [Indexed: 12/23/2022] Open
Abstract
Background Cellular processes occur within dynamic and multi-molecular compartments whose characterization requires analysis at high spatio-temporal resolution. Notable examples for such complexes are cell-matrix adhesion sites, consisting of numerous cytoskeletal and signaling proteins. These adhesions are highly variable in their morphology, dynamics, and apparent function, yet their molecular diversity is poorly defined. Methodology/Principal Findings We present here a compositional imaging approach for the analysis and display of multi-component compositions. This methodology is based on microscopy-acquired multicolor data, multi-dimensional clustering of pixels according to their composition similarity and display of the cellular distribution of these composition clusters. We apply this approach for resolving the molecular complexes associated with focal-adhesions, and the time-dependent effects of Rho-kinase inhibition. We show here compositional variations between adhesion sites, as well as ordered variations along the axis of individual focal-adhesions. The multicolor clustering approach also reveals distinct sensitivities of different focal-adhesion-associated complexes to Rho-kinase inhibition. Conclusions/Significance Multicolor compositional imaging resolves “molecular signatures” characteristic to focal-adhesions and related structures, as well as sub-domains within these adhesion sites. This analysis enhances the spatial information with additional “contents-resolved” dimensions. We propose that compositional imaging can serve as a powerful tool for studying complex multi-molecular assemblies in cells and for mapping their distribution at sub-micron resolution.
Collapse
Affiliation(s)
- Eli Zamir
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| | - Zvi Kam
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
425
|
The evolution of tools for protein phosphorylation site analysis: from discovery to clinical application. Biotechniques 2008; 44:671-9. [DOI: 10.2144/000112800] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The importance of the analysis of signaling pathways has been proven for many years by the elucidation of key signaling molecules. However, in most cases these pathways tend to represent a rather narrow view of the biological state under investigation. Clearly a more detailed understanding of the complexities of cross-talk between signaling pathways is required to further our knowledge of normal and disease processes. The tools that provide the framework for this increased understanding of biology, those that enable identification, characterization, and quantitation of sites of phosphorylation in proteins, have advanced over the past 25 years. This review will present a brief overview of the history of the tools used in phosphorylation analysis and the latest technologies that are being applied in this field, such as mass spectrometry (for broad-based discovery efforts) and flow cytometry (for translation to clinical applications).
Collapse
|
426
|
Ross D, Hughes T. Current and emerging tests for the laboratory monitoring of chronic myeloid leukaemia and related disorders. Pathology 2008; 40:231-46. [DOI: 10.1080/00313020801916172] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
427
|
TNF-induced MAP kinase activation oscillates in time. Biochem Biophys Res Commun 2008; 371:906-11. [PMID: 18384751 DOI: 10.1016/j.bbrc.2008.03.113] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 03/20/2008] [Indexed: 01/17/2023]
Abstract
Oscillations in the activation of multiple signaling pathways have never been shown before. Our results presented in the previous accompanying paper showed that TNF induces highly dynamic oscillations in mRNA production in approximately 13% of the mouse genome. Here, we further analyze the TNF time-series microarray data and find that multiple signaling components downstream of the TNF receptor undergo oscillations. Prior studies implicate IkappaBalpha and A-20 as the only oscillatory components in the TNF signaling cascade. We find however, that other components, such as TRAF1, displayed oscillations. This suggested the possibility that all signaling output from the TNF receptor may be oscillatory in nature. Indeed, we show that TNF triggers oscillations in the phosphorylation of three MAP kinases, as well as p65. Because Baltimore and colleagues had proposed that NF-kappaB drives the oscillatory nature of the IkappaBalpha/NF-kappaB feedback loop, we studied the effects of an NF-kappaB super-repressor on oscillations in MAPK phosphorylation; we find that the super-repressor altered the amplitude and frequency of MAP kinase phosphorylation, but failed to abolish oscillations. These results attest to a role for NF-kappaB as a modulator, but not the sole determinant of cyclical activation of signal transduction pathways. These results, together with those of the two accompanying papers, constitute a new paradigm through which cells orchestrate signaling molecules to produce highly dynamic physiological processes such as gene transcription and protein secretion. In view of the discovery that multiple phosphorylation pathways display dynamic oscillations, time-resolved, instead of static, measurements of kinase phosphorylation should become the experimental norm.
Collapse
|
428
|
Pawson T, Linding R. Network medicine. FEBS Lett 2008; 582:1266-70. [PMID: 18282479 DOI: 10.1016/j.febslet.2008.02.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2008] [Revised: 02/09/2008] [Accepted: 02/11/2008] [Indexed: 12/20/2022]
Abstract
To more effectively target complex diseases like cancer, diabetes and schizophrenia, we may need to rethink our strategies for drug development and the selection of molecular targets for pharmacological treatments. Here, we discuss the potential use of protein signaling networks as the targets for new therapeutic intervention. We argue that by targeting the architecture of aberrant signaling networks associated with cancer and other diseases new therapeutic strategies can be implemented. Transforming medicine into a network driven endeavour will require quantitative measurements of cell signaling processes; we will describe how this may be performed and combined with new algorithms to predict the trajectories taken by a cellular system either in time or through disease states. We term this approach, network medicine.
Collapse
Affiliation(s)
- Tony Pawson
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Canada.
| | | |
Collapse
|
429
|
Single-cell, phosphoepitope-specific analysis demonstrates cell type- and pathway-specific dysregulation of Jak/STAT and MAPK signaling associated with in vivo human immunodeficiency virus type 1 infection. J Virol 2008; 82:3702-12. [PMID: 18216116 DOI: 10.1128/jvi.01582-07] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Despite extensive evidence of cell signaling alterations induced by human immunodeficiency virus type 1 (HIV-1) in vitro, the relevance of these changes to the clinical and/or immunologic status of HIV-1-infected individuals is often unclear. As such, mapping the details of cell type-specific degradation of immune function as a consequence of changes to signaling network responses has not been readily accessible. We used a flow cytometric-based assay of signaling to determine Janus kinase/signal transducers and activators of transcription (Jak/STAT) signaling changes at the single-cell level within distinct cell subsets from the primary immune cells of HIV-1-infected donors. We identified a specific defect in granulocyte-macrophage colony-stimulating factor (GM-CSF)-driven Stat5 phosphorylation in the monocytes of HIV-1+ donors. This inhibition was statistically significant in a cohort of treated and untreated individuals. Ex vivo Stat5 phosphorylation levels varied among HIV-1+ donors but did not correlate with CD4(+) T-cell counts or HIV-1 plasma viral load. Low Stat5 activation occurred in HIV-1-infected donors despite normal GM-CSF receptor levels. Investigation of mitogen-activated protein kinase (MAPK) pathways, also stimulated by GM-CSF, led to the observation that lipopolysaccharide-stimulated extracellular signal-regulated kinase phosphorylation is enhanced in monocytes. Thus, we have identified a specific, imbalanced monocyte signaling profile, with inhibition of STAT and enhancement of MAPK signaling, associated with HIV-1 infection. This understanding of altered monocyte signaling responses that contribute to defective antigen presentation during HIV-1 infection could lead to immunotherapeutic approaches that compensate for the deficiency.
Collapse
|
430
|
Abolition of stress-induced protein synthesis sensitizes leukemia cells to anthracycline-induced death. Blood 2008; 111:2866-77. [PMID: 18182573 DOI: 10.1182/blood-2007-07-103242] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anthracycline action has been thought to involve the neosynthesis of proapoptotic gene products and to therefore depend on protein synthesis for optimal effect. We found that inhibition of general, but not rapamycin-sensitive (cap-dependent), protein synthesis in the preapoptotic period enhanced anthracycline-induced acute myelogenous leukemia (AML) cell death, both in vitro and in several animal AML models. Pre-apoptotic anthracycline-exposed AML cells had altered translational specificity, with enhanced synthesis of a subset of proteins, including endoplasmatic reticulum chaperones. The altered translational specificity could be explained by perturbation (protein degradation, truncation, or dephosphorylation) of the cap-dependent translation initiation machinery and of proteins control-ing translation of specific mRNAs. We propose that judiciously timed inhibition of cap-independent translation is considered for combination therapy with anthracyclines in AML.
Collapse
|
431
|
Abstract
Hematopoietic stem cells (HSC) play critical roles in maintaining blood cell production for the lifetime of the organism. Considerable progress has been made in their isolation from mouse bone marrow to high levels of purity based on a combination of cell-surface phenotype and functional characteristics. In addition, in vitro assays have been established that provide important tools for study of hematopoietic differentiation from HSC and for differentiation to generate HSC from embryonic stem cells. Although these in vitro studies provide a window on the temporal function and differentiation of HSC progeny, the transplantation assay still serves as the gold standard for quantitative and qualitative analysis of murine HSC biology. There are now many flavors of syngeneic and xenogeneic HSC transplant, all focused on quantitative assessment of repopulating function. As a vehicle for genetic modification of HSC, retroviral-mediated gene transfer followed by transplantation has had a major impact upon our understanding of genetic disorders, gene therapy, and leukemogenesis. This overview chapter summarizes the growing number of tools available for HSC research and specifically ties together the methods in chapters of the second edition of Hematopoietic Stem Cell Protocols.
Collapse
Affiliation(s)
- William Tse
- Department of Medicine, Division of Hematology-Oncology, Center for Stem Cell and Regenerative Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|
432
|
Ornatsky OI, Kinach R, Bandura DR, Lou X, Tanner SD, Baranov VI, Nitz M, Winnik MA. Development of analytical methods for multiplex bio-assay with inductively coupled plasma mass spectrometry. JOURNAL OF ANALYTICAL ATOMIC SPECTROMETRY 2008; 23:463-469. [PMID: 19122859 PMCID: PMC2600572 DOI: 10.1039/b710510j] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Advances in the development of highly multiplexed bio-analytical assays with inductively coupled plasma mass spectrometry (ICP-MS) detection are discussed. Use of novel reagents specifically designed for immunological methods utilizing elemental analysis is presented. The major steps of method development, including selection of elements for tags, validation of tagged reagents, and examples of multiplexed assays, are considered in detail. The paper further describes experimental protocols for elemental tagging of antibodies, immunostaining of live and fixed human leukemia cells, and preparation of samples for ICP-MS analysis. Quantitative analysis of surface antigens on model cell lines using a cocktail of seven lanthanide labeled antibodies demonstrated high specificity and concordance with conventional immunophenotyping.
Collapse
Affiliation(s)
- Olga I Ornatsky
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Room 407, 164 College Street, Toronto, Ontario, Canada, M5S 3G9
| | | | | | | | | | | | | | | |
Collapse
|
433
|
|
434
|
High-content single-cell drug screening with phosphospecific flow cytometry. Nat Chem Biol 2007; 4:132-42. [DOI: 10.1038/nchembio.2007.59] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Accepted: 10/30/2007] [Indexed: 11/08/2022]
|
435
|
|
436
|
Capturing cell-fate decisions from the molecular signatures of a receptor-dependent signaling response. Mol Syst Biol 2007; 3:150. [PMID: 18059445 PMCID: PMC2174630 DOI: 10.1038/msb4100197] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 10/25/2007] [Indexed: 12/25/2022] Open
Abstract
We examined responses of the B-cell antigen receptor-dependent intracellular signaling network to targeted perturbations induced through siRNA-mediated depletion of select signaling intermediates. The constituent nodes displayed graded sensitivities, which resulted from the differential effects of perturbations on the kinetic and quantitative aspects of phosphorylation at each node. By taking the rate of initial phosphorylation, rate of subsequent dephosphorylation, and the total intensity of phosphorylation at each node as separate signaling parameters, we generated data-driven models that accurately predicted the cellular responses of apoptosis, proliferation, and cytokine secretion. Importantly, the effects of perturbation on the primary target alone did not yield successful models. Rather, it also required incorporation of secondary effects on many other nodes. A significant feature of these models was that the three signaling parameters derived from each node functioned largely as independent entities, making distinctive contributions to the cellular response. Thus, the kinetic and quantitative features of phosphorylation at a node appear to play discrete roles during signal processing.
Collapse
|
437
|
Anensen N, Oyan AM, Bourdon JC, Kalland KH, Bruserud O, Gjertsen BT. A distinct p53 protein isoform signature reflects the onset of induction chemotherapy for acute myeloid leukemia. Clin Cancer Res 2007; 12:3985-92. [PMID: 16818696 DOI: 10.1158/1078-0432.ccr-05-1970] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE The antioncogene protein product p53 has not been studied previously in cancer patients during in vivo chemotherapy. This study examined the early p53 protein and gene expression during induction chemotherapy in acute myeloid leukemia (AML). EXPERIMENTAL DESIGN Leukemic cells were collected from five AML patients during their first 18 hours of induction chemotherapy and examined for p53 protein and gene expression by one- and two-dimensional gel immunoblot and high-density gene expression arrays. RESULTS Up-regulation of p53 protein expression was detected in AML patients posttreatment in vivo. One- and two-dimensional gel immunoblots showed two main forms of p53, denominated alpha p53 and delta p53, both recognized by various NH2-terminal directed antibodies. As a response to treatment, we detected rapid accumulation of alpha p53, with significantly altered protein expression levels already after 2 hours. The accumulation of alpha p53 was accompanied by increased transcription of putative p53 target genes and subsequent cytopenia in the patients. CONCLUSION Up-regulation of the p53 protein and target genes seems to be a prominent feature in induction chemotherapy of AML. The rapid shift from a shorter p53 protein form (delta) toward the full-length protein (alpha) underscores the complexity of p53 protein modulation in patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Nina Anensen
- Hematology Section, Institute of Medicine, University of Bergen, Norway
| | | | | | | | | | | |
Collapse
|
438
|
Rechavi O, Goldstein I, Vernitsky H, Rotblat B, Kloog Y. Intercellular transfer of oncogenic H-Ras at the immunological synapse. PLoS One 2007; 2:e1204. [PMID: 18030338 PMCID: PMC2065899 DOI: 10.1371/journal.pone.0001204] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Accepted: 10/28/2007] [Indexed: 12/05/2022] Open
Abstract
Immune cells establish dynamic adhesive cell–cell interactions at a specific contact region, termed the immunological synapse (IS). Intriguing features of the IS are the formation of regions of plasma membrane fusion and the intercellular exchange of membrane fragments between the conjugated cells. It is not known whether upon IS formation, intact intracellular proteins can transfer from target cells to lymphocytes to allow the transmission of signals across cell boundaries. Here we show by both FACS and confocal microscopy that human lymphocytes acquire from the cells they scan the inner-membrane protein H-Ras, a G-protein vital for common lymphocyte functions and a prominent participant in human cancer. The transfer was cell contact-dependent and occurred in the context of cell-conjugate formation. Moreover, the acquisition of oncogenic H-RasG12V by natural killer (NK) and T lymphocytes had important biological functions in the adopting lymphocytes: the transferred H-RasG12V induced ERK phosphorylation, increased interferon-γ and tumor necrosis factor-α secretion, enhanced lymphocyte proliferation, and augmented NK-mediated target cell killing. Our findings reveal a novel mode of cell-to-cell communication—allowing lymphocytes to extend the confines of their own proteome—which may moreover play an important role in natural tumor immunity.
Collapse
Affiliation(s)
- Oded Rechavi
- Department of Neurobiochemistry, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Itamar Goldstein
- Immunology Program, Cancer Research Center, Chaim Sheba Medical Center, Tel Hashomer and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * To whom correspondence should be addressed. E-mail: (IG); (YK)
| | - Helly Vernitsky
- Immunology Program, Cancer Research Center, Chaim Sheba Medical Center, Tel Hashomer and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Barak Rotblat
- Department of Neurobiochemistry, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Yoel Kloog
- Department of Neurobiochemistry, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
- * To whom correspondence should be addressed. E-mail: (IG); (YK)
| |
Collapse
|
439
|
Shapiro HM. Cytometry in monoclonal B-cell lymphocytosis and chronic lymphocytic leukaemia--the Hunting of the Snark? Br J Haematol 2007; 139:772-3. [PMID: 18021090 DOI: 10.1111/j.1365-2141.2007.06855.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cytometry has become important in the detection and determination of risk of monoclonal B-cell lymphocytosis; methodology has changed, and will continue to change, as cytometric technology changes.
Collapse
|
440
|
Abstract
Abnormalities of cytokine and growth factor signaling pathways are characteristic of all forms of leukemia: lymphoid and myeloid, acute and chronic. In normal hematopoietic cells, cytokines provide the stimulus for proliferation, survival, self-renewal, differentiation and functional activation. In leukemic cells, these pathways are usurped to subserve critical parts of the malignant program. In this review, our current knowledge of leukemic cell cytokine signaling will be summarized, and some speculations on the significance and implications of these insights will be advanced. A better understanding of aberrant cytokine signaling in leukemia should provide additional targets for the rational therapy of these diseases.
Collapse
Affiliation(s)
- R A Van Etten
- Molecular Oncology Research Institute and Division of Hematology/Oncology, Tufts-New England Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
441
|
Vakkila J, Nieminen U, Siitonen S, Turunen U, Halme L, Nuutinen H, Mustonen H, Puolakkainen P, Färkkilä M, Repo H. A novel modification of a flow cytometric assay of phosphorylated STAT1 in whole blood monocytes for immunomonitoring of patients on IFN alpha regimen. Scand J Immunol 2007; 67:95-102. [PMID: 18028288 DOI: 10.1111/j.1365-3083.2007.02028.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We explored whether episodes stimulating leucocytes in vivo could be tracked from whole blood samples by monitoring activation of STAT1 by flow cytometry. The method was tested in hepatitis C patients (n = 9) that were on interferon (IFN)alpha regimen. CD14+ monocytes responded strongly to IFNalpha/gamma being sensitive indicators for recent immune activation. At 45 min after s.c. IFNalpha 91% of monocytes were phosphorylated STAT1+. The frequency of responding cells decreased to a base level within 6 h. Monocytes, however, had a long-term deficient phosphorylated STAT1 response to IFNalphain vitro that in patients on standard IFNalpha regimen lasted for 48 h. In patients on pegylated IFNalpha the phosphorylated STAT1 response was completely absent. We conclude that whole blood analysis of STAT1 activation by flow cytometry is applicable to monitor immune cells in patient material.
Collapse
Affiliation(s)
- J Vakkila
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
442
|
Sklar LA, Carter MB, Edwards BS. Flow cytometry for drug discovery, receptor pharmacology and high-throughput screening. Curr Opin Pharmacol 2007; 7:527-34. [PMID: 17652026 PMCID: PMC2230635 DOI: 10.1016/j.coph.2007.06.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 06/06/2007] [Accepted: 06/15/2007] [Indexed: 12/16/2022]
Abstract
Although flow cytometry is viewed as a mature technology, there have been dramatic advances in analysis capabilities, sorting, sample handling and sensitivity in the past decade. These advances contribute to its application in biological and chemical diversity, sample throughput, high content, and complex systems biology. This article will evaluate the new opportunities for flow cytometry relating to receptor assembly and pharmacology, as well as a range of screening applications.
Collapse
Affiliation(s)
- Larry A Sklar
- The New Mexico Molecular Libraries Screening Center, Cancer Research and Treatment Center, The University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | | | | |
Collapse
|
443
|
Abstract
Proteomics technologies are emerging as a useful tool in the identification of disease biomarkers, and in defining and characterising both normal physiological and disease processes. Many cellular changes in protein expression in response to an external stimulus or mutation can only be characterised at the proteome level. In these cases protein expression is often controlled by altered rates of translation and/or degradation, making proteomics an important tool in the analysis of biological systems. In the leukaemias, post-translational modification of proteins (e.g. phosphorylation, acetylation) plays a key role in the molecular pathology of the disease: such modifications can now be detected with novel proteomic methods. In a clinical setting, serum remains a relatively un-mined source of information for prognosis and response to therapy. This protein rich fluid represents an opportunity for proteomics research to benefit hematologists and others. In this review, we discuss the technologies available for the study of the proteome that offer realistic opportunities in haematology.
Collapse
Affiliation(s)
- Richard D Unwin
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Medical and Human Sciences, University of Manchester, Christie Hospital, Kinnaird House, Kinnaird Road, Withington, Manchester, UK M20 4QL.
| | | |
Collapse
|
444
|
Bowers RK, Marder P, Green LJ, Horn CL, Faber AL, Thomas JE. A platelet biomarker for assessing phosphoinositide 3-kinase inhibition during cancer chemotherapy. Mol Cancer Ther 2007; 6:2600-7. [PMID: 17766838 DOI: 10.1158/1535-7163.mct-06-0746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Thrombin cleavages of selective proteinase-activated receptors (PAR) as well as PAR-activating peptide ligands can initiate the phosphoinositide 3-kinase (PI3K) signaling cascade in platelets. Downstream to this event, fibrinogen receptors on platelets undergo conformational changes that enhance fibrinogen binding. In our study, we used this phenomenon as a surrogate biomarker for assessing effects on PI3K activity. Our method, using flow cytometric measurement of fluorescent ligand and antibody binding, uncovered a 16- to 45-fold signal window after PAR-induced platelet activation. Pretreatment (in vitro) with the PI3K inhibitors wortmannin and LY294002 resulted in concentration-dependent inhibition at predicted potencies. In addition, platelets taken from mice treated with wortmannin were blocked from PAR-induced ex vivo activation concomitantly with a decrease in phosphorylation of AKT from excised tumor xenografts. This surrogate biomarker assay was successfully tested (in vitro) on blood specimens received from volunteer cancer patients. Our results indicate that measurement of platelet activation could serve as an effective drug activity biomarker during clinical evaluation of putative PI3K inhibitors.
Collapse
Affiliation(s)
- Rita K Bowers
- Lilly Research Laboratories, Indianapolis, Indiana, USA .
| | | | | | | | | | | |
Collapse
|
445
|
Yan GR, He QY. Functional proteomics to identify critical proteins in signal transduction pathways. Amino Acids 2007; 35:267-74. [PMID: 17704892 DOI: 10.1007/s00726-007-0594-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Accepted: 05/25/2007] [Indexed: 01/07/2023]
Abstract
Reversible protein phosphorylation plays a crucial role in the regulation of signaling pathways that control various biological responses, such as cell growth, differentiation, invasion, metastasis and apoptosis. Proteomics is a powerful research approach for fully monitoring global molecular responses to the activation of signal transduction pathways. Identification of different phosphoproteins and their phosphorylation sites by functional proteomics provides informational insights into signaling pathways triggered by all kinds of factors. This review summarizes how functional proteomics can be used to answer specific questions related to signal transduction systems of interest. By examining our own example on identifying the novel phosphoproteins in signaling pathways activated by EB virus-encoded latent membrane protein 1 (LMP1), we demonstrated a functional proteomic strategy to elucidate the molecular activity of phosphorylated annexin A2 in LMP1 signaling pathway. Functional profiling of signaling pathways is promising for the identification of novel targets for drug discovery and for the understanding of disease pathogenesis.
Collapse
Affiliation(s)
- G-R Yan
- Institutes of Life and Health Engineering, Jinan University, Guangzhou, China
| | | |
Collapse
|
446
|
Jang DJ, Guo M, Wang D. Proteomic and biochemical studies of calcium- and phosphorylation-dependent calmodulin complexes in Mammalian cells. J Proteome Res 2007; 6:3718-28. [PMID: 17696464 DOI: 10.1021/pr0703268] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein conformational changes due to cofactor binding (e.g., metal ions, heme) and/or post-translational modifications (e.g., phosphorylation) modulate dynamic protein complexes. Calmodulin (CaM) plays an essential role in regulating calcium signaling and homeostasis. Herein, we report a straightforward and systematic approach to identify potential calcium- and phosphorylation-dependent CaM complexes in a proteome-wide manner. We have identified over 120 CaM-associated proteins encompassing four different classes of CaM binding in HeLa cells, namely, calcium-dependent and phosphorylation-dependent (e.g., EDD1), calcium-dependent and phosphorylation-independent (e.g., myosin IE), calcium-independent and phosphorylation-dependent (e.g., DDX3), and calcium-independent and phosphorylation-independent (e.g., DDX5). To demonstrate the utility of our method in understanding biological pathways, we showed that in vivo phosphorylation of inositol 1,4,5-triphosphate receptor type 1 (IP3R1) at Ser1598 significantly reduced the affinity of its Ca2+-dependent CaM binding. However, phosphorylation of IP3R1 did not substantially affect its Ca2+-independent CaM binding. These results shed new lights on the mechanism underlying the marked increase of Ca2+ release due to IP3R1 phosphorylation. We further showed that staurosporine-sensitive kinase(s) and phosphatase PP1 play a critical role in modulating the phosphorylation-dependent CaM binding of IP3R1. Our method may serve as a general strategy to identify and characterize phosphorylation-dependent protein complexes, to pinpoint the phosphorylation sites and associated kinase(s) and phosphatase(s) involved in the protein-protein interactions, and to functionally characterize these complexes in mammalian cells.
Collapse
Affiliation(s)
- Deok-Jin Jang
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, MS 84-171, Berkeley, California 94720, USA
| | | | | |
Collapse
|
447
|
Abstract
RNAi screening in mammalian cells has become a valuable method to identify and describe genetic relationships in both basic biology and disease mechanisms. Multiple efforts are underway to standardize how RNAi screening data are reported, including establishing experimental criteria for defining a validated hit from a screen, and the extent to which the primary screening data themselves are reported. These discussions have identified several key areas that require consistency, or at least understanding, before RNAi screening data can be used generally. Successfully addressing these targeted areas would broaden the use of RNAi screening data beyond advancing one or a few hits into validation experiments, to enable verification of primary screening data, and to facilitate comparisons between sample groups based on screening profiles. Areas for improving RNAi screening include general guidelines for validating hits from screens, the creation of standardized reporting structures for RNAi screening data, such as Minimum Information About an RNAi Experiment (MIARE), statistical methods for analyzing screening data that explicitly account for differences between screening RNAi reagents versus small molecules, and technical improvements to RNAi screening that improve the analysis of gene knockdowns, including multiparametric approaches, such as high-content screening. This review will discuss how these approaches can improve RNAi screening data at the community level and for an individual researcher trying to manage an RNAi screen.
Collapse
Affiliation(s)
- Steven A Haney
- Department of Biological Technologies, Wyeth Research, 35 Cambridge Park Drive, Cambridge, MA 02140, USA.
| |
Collapse
|
448
|
Miller-Jensen K, Janes KA, Brugge JS, Lauffenburger DA. Common effector processing mediates cell-specific responses to stimuli. Nature 2007; 448:604-8. [PMID: 17637676 DOI: 10.1038/nature06001] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 06/07/2007] [Indexed: 11/09/2022]
Abstract
The fundamental components of many signalling pathways are common to all cells. However, stimulating or perturbing the intracellular network often causes distinct phenotypes that are specific to a given cell type. This 'cell specificity' presents a challenge in understanding how intracellular networks regulate cell behaviour and an obstacle to developing drugs that treat signalling dysfunctions. Here we apply a systems-modelling approach to investigate how cell-specific signalling events are integrated through effector proteins to cause cell-specific outcomes. We focus on the synergy between tumour necrosis factor and an adenoviral vector as a therapeutically relevant stimulus that induces cell-specific responses. By constructing models that estimate how kinase-signalling events are processed into phenotypes through effector substrates, we find that accurate predictions of cell specificity are possible when different cell types share a common 'effector-processing' mechanism. Partial-least-squares regression models based on common effector processing accurately predict cell-specific apoptosis, chemokine release, gene induction, and drug sensitivity across divergent epithelial cell lines. We conclude that cell specificity originates from the differential activation of kinases and other upstream transducers, which together enable different cell types to use common effectors to generate diverse outcomes. The common processing of network signals by downstream effectors points towards an important cell biological principle, which can be applied to the understanding of cell-specific responses to targeted drug therapies.
Collapse
Affiliation(s)
- Kathryn Miller-Jensen
- Center for Cell Decision Processes, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
449
|
Shachaf CM, Perez OD, Youssef S, Fan AC, Elchuri S, Goldstein MJ, Shirer AE, Sharpe O, Chen J, Mitchell DJ, Chang M, Nolan GP, Steinman L, Felsher DW. Inhibition of HMGcoA reductase by atorvastatin prevents and reverses MYC-induced lymphomagenesis. Blood 2007; 110:2674-84. [PMID: 17622571 PMCID: PMC1988916 DOI: 10.1182/blood-2006-09-048033] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Statins are a class of drugs that inhibit 3-hydroxy-3-methylglutaryl-coenzyme A (HMGcoA) reductase, a critical enzyme in the mevalonate pathway. Several reports document that statins may prevent different human cancers. However, whether or not statins can prevent cancer is controversial due to discordant results. One possible explanation for these conflicting conclusions is that only some tumors or specific statins may be effective. Here, we demonstrate in an in vivo transgenic model in which atorvastatin reverses and prevents the onset of MYC-induced lymphomagenesis, but fails to reverse or prevent tumorigenesis in the presence of constitutively activated K-Ras (G12D). Using phosphoprotein fluorescence-activated cell sorter (FACS) analysis, atorvastatin treatment was found to result in the inactivation of the Ras and ERK1/2 signaling pathways associated with the dephosphorylation and inactivation of MYC. Correspondingly, tumors with a constitutively activated K-Ras (G12D) did not exhibit dephosphorylation of ERK1/2 and MYC. Atorvastatin's effects on MYC were specific to the inhibition of HMGcoA reductase, as treatment with mevalonate, the product of HMG-CoA reductase activity, abrogated these effects and inhibited the ability of atorvastatin to reverse or suppress tumorigenesis. Also, RNAi directed at HMGcoA reductase was sufficient to abrogate the neoplastic properties of MYC-induced tumors. Thus, atorvastatin, by inhibiting HMGcoA reductase, induces changes in phosphoprotein signaling that in turn prevent MYC-induced lymphomagenesis.
Collapse
Affiliation(s)
- Catherine M Shachaf
- Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, CA 94305-5151, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
450
|
McLornan DP, McMullin MF, Johnston P, Longley DB. Molecular mechanisms of drug resistance in acute myeloid leukaemia. Expert Opin Drug Metab Toxicol 2007; 3:363-77. [PMID: 17539744 DOI: 10.1517/17425255.3.3.363] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Resistance to chemotherapy in acute myeloid leukaemia is a major obstacle to a successful outcome for many patients. Often, there is resistance against a broad range of drugs due to multiple, simultaneously active processes. These mechanisms include effects on drug influx and efflux, drug activation/inactivation, DNA repair mechanisms, altered response of end targets, an altered haematopoietic microenvironment and dysfunctional apoptotic pathways. This article reviews the factors that determine leukaemic cell chemosensitivity and discusses the potential for rationally guided therapy.
Collapse
Affiliation(s)
- Donal P McLornan
- Medical Research Council Clinical Research Fellow, Queen's University Belfast, Centre for Cancer Research and Cell Biology, BT7 1NN, Northern Ireland, UK
| | | | | | | |
Collapse
|