1
|
Executable pathway analysis using ensemble discrete-state modeling for large-scale data. PLoS Comput Biol 2019; 15:e1007317. [PMID: 31479446 PMCID: PMC6743792 DOI: 10.1371/journal.pcbi.1007317] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 09/13/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022] Open
Abstract
Pathway analysis is widely used to gain mechanistic insights from high-throughput omics data. However, most existing methods do not consider signal integration represented by pathway topology, resulting in enrichment of convergent pathways when downstream genes are modulated. Incorporation of signal flow and integration in pathway analysis could rank the pathways based on modulation in key regulatory genes. This implementation can be facilitated for large-scale data by discrete state network modeling due to simplicity in parameterization. Here, we model cellular heterogeneity using discrete state dynamics and measure pathway activities in cross-sectional data. We introduce a new algorithm, Boolean Omics Network Invariant-Time Analysis (BONITA), for signal propagation, signal integration, and pathway analysis. Our signal propagation approach models heterogeneity in transcriptomic data as arising from intercellular heterogeneity rather than intracellular stochasticity, and propagates binary signals repeatedly across networks. Logic rules defining signal integration are inferred by genetic algorithm and are refined by local search. The rules determine the impact of each node in a pathway, which is used to score the probability of the pathway's modulation by chance. We have comprehensively tested BONITA for application to transcriptomics data from translational studies. Comparison with state-of-the-art pathway analysis methods shows that BONITA has higher sensitivity at lower levels of source node modulation and similar sensitivity at higher levels of source node modulation. Application of BONITA pathway analysis to previously validated RNA-sequencing studies identifies additional relevant pathways in in-vitro human cell line experiments and in-vivo infant studies. Additionally, BONITA successfully detected modulation of disease specific pathways when comparing relevant RNA-sequencing data with healthy controls. Most interestingly, the two highest impact score nodes identified by BONITA included known drug targets. Thus, BONITA is a powerful approach to prioritize not only pathways but also specific mechanistic role of genes compared to existing methods. BONITA is available at: https://github.com/thakar-lab/BONITA.
Collapse
|
2
|
Zhong C, Song H, Weiss A, Tan WH, Coury S, Huang J. Myofibromatosis presenting as reticulated vascular changes and subcutaneous atrophy in a patient with somatic mosaicism of PDGFRB
mutation. Br J Dermatol 2018; 179:1408-1409. [DOI: 10.1111/bjd.16995] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- C.S. Zhong
- Harvard Medical School; 50 Staniford Street 2nd Floor Boston MA 02114 U.S.A
| | - H. Song
- Harvard Medical School; 50 Staniford Street 2nd Floor Boston MA 02114 U.S.A
| | - A. Weiss
- Department of Pediatric Hematology/Oncology; Maine Medical Center; Scarborough ME U.S.A
| | - W.-H. Tan
- Division of Genetics and Genomics; Boston Children's Hospital; Boston MA U.S.A
| | - S. Coury
- Division of Genetics and Genomics; Boston Children's Hospital; Boston MA U.S.A
| | - J.T. Huang
- Department of Immunology; Dermatology Program, Boston Children's Hospital; Boston MA U.S.A
| |
Collapse
|
3
|
Tseng T, Uen W, Tseng J, Lee S. Enhanced chemosensitization of anoikis-resistant melanoma cells through syndecan-2 upregulation upon anchorage independency. Oncotarget 2017; 8:61528-61537. [PMID: 28977882 PMCID: PMC5617442 DOI: 10.18632/oncotarget.18616] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 05/29/2017] [Indexed: 01/05/2023] Open
Abstract
Syndecan family proteins are heparan sulfate proteoglycans, which involved in various cellular activities and associating with metastatic potential and chemosensitivity of tumor cells. Melanoma is one of malignant tumors with poor prognosis upon metastasis. Previously, we had shown that melanoma cells remained survived under cell detachment, which was similar to the initial steps of tumor metastasis. Downregulation of syndecan-1 and upregulation of syndecan-2 in melanoma A375 cells were observed by different suspension conditions. Specific gene alterations also increased melanoma malignancy under anchorage independency. Thus, we would like to investigate in further the role of specific gene alteration, so that it could be used to develop novel strategy to treat melanoma. In this paper, we found that syndecan-2 expression level as well the kinase phosphorylation levels increased upon anchorage independency. The pathway to regulate syndecan-2 expression shifted from PKCα/β-dependent under adhesion into PKCδ-dependent under cell suspension. Manipulation of syndecan-2 expression showed that PI3K and ERK phosphorylation as well the migratory ability increased with increased syndecan-2 expression level. In addition, suspended melanoma cells were more sensitive to chemoagents, which correlated with syndecan-2 overexpression, PI3K and ERK activations, serum level, and the presence of glycosaminoglycans. In conclusion, we showed upregulation of syndecan-2 in anoikis-resistant melanoma cells enhanced chemosensitivity through PI3K and ERK activation. This observation would support and refine the strategy of adjuvant chemotherapy to overcome metastatic melanoma.
Collapse
Affiliation(s)
- TingTing Tseng
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - WuChing Uen
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.,Department of Hematology and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei City 111, Taiwan
| | - JenChih Tseng
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - ShaoChen Lee
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
4
|
Abstract
The epidermal growth factor receptor (EGFR) is one of the most well-studied signaling pathways in cancer progression. As a result, numerous therapeutics including small-molecule inhibitors and monoclonal antibodies have been developed to target this critical oncogenic driver. Several of these EGFR inhibitors (EGFRi) have been evaluated in metastatic breast cancer, as high-level EGFR expression in primary tumors correlates with the highly aggressive basal-like phenotype and predicts for poor patient prognosis. Surprisingly, these trials have been unanimously unsuccessful at improving patient outcomes. Numerous factors, such as lack of proper patient selection may have contributed to the failure of these trials. However, recent findings suggest that there are fundamental changes in EGFR signaling that take place during primary tumor invasion, dissemination and ultimate metastasis of breast cancer cells. Herein, we review the outcomes of EGFR-targeted clinical trials in breast cancer and explore our current understanding of EGFR signaling within primary mammary tumors and how these events are altered in the metastatic setting. Overall, we put forth the hypothesis that fundamental changes in EGFR signaling between primary and metastatic tumors, a process we term the 'EGFR paradox,' contribute to the clinically observed inherent resistance to EGFRi. Furthermore, this hypothesis introduces the possibility of utilizing EGFR agonism as a potential therapeutic approach for the treatment of metastatic breast cancer.
Collapse
|
5
|
Vascular Endothelial Cell Growth Factor A Acts via Platelet-Derived Growth Factor Receptor α To Promote Viability of Cells Enduring Hypoxia. Mol Cell Biol 2016; 36:2314-27. [PMID: 27325673 DOI: 10.1128/mcb.01019-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 06/07/2016] [Indexed: 12/20/2022] Open
Abstract
Vascular endothelial cell growth factor A (VEGF) is a biologically and therapeutically important growth factor because it promotes angiogenesis in response to hypoxia, which underlies a wide variety of both physiological and pathological settings. We report here that both VEGF receptor 2 (VEGFR2)-positive and -negative cells depended on VEGF to endure hypoxia. VEGF enhanced the viability of platelet-derived growth factor receptor α (PDGFRα)-positive and VEGFR2-negative cells by enabling indirect activation of PDGFRα, thereby reducing the level of p53. We conclude that the breadth of VEGF's influence extends beyond VEGFR-positive cells and propose a plausible mechanistic explanation of this phenomenon.
Collapse
|
6
|
Ilyas SI, Mertens JC, Bronk SF, Hirsova P, Dai H, Roberts LR, Kaufmann SH, Gores GJ. Platelet-derived growth factor primes cancer-associated fibroblasts for apoptosis. J Biol Chem 2014; 289:22835-22849. [PMID: 24973208 PMCID: PMC4132787 DOI: 10.1074/jbc.m114.563064] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/03/2014] [Indexed: 12/19/2022] Open
Abstract
Desmoplastic malignancies such as cholangiocarcinoma (CCA) are characterized by a dense stroma containing an abundance of myofibroblasts termed cancer-associated fibroblasts (CAF). The CAF phenotype represents an "activated state" in which cells are primed for cell death triggered by BH3 mimetics. Accordingly, this primed state may be therapeutically exploited. To elucidate the mechanisms underlying this poorly understood apoptotic priming, we examined the role of platelet-derived growth factor (PDGF) in CAF priming for cell death given its prominent role in CAF activation. PDGF isomers PDGF-B and PDGF-D are abundantly expressed in CCA cells derived from human specimens. Either isomer sensitizes myofibroblasts to cell death triggered by BH3 mimetics. Similar apoptotic sensitization was observed with co-culture of myofibroblasts and CCA cells. Profiling of Bcl-2 proteins expressed by PDGF-primed myofibroblasts demonstrated an increase in cellular levels of Puma. PDGF-mediated increases in cellular Puma levels induced proapoptotic changes in Bak, which resulted in its binding to Bcl-2. Short hairpin RNA-mediated down-regulation of Puma conferred resistance to PDGF-mediated apoptotic priming. Conversely, the BH3 mimetic navitoclax disrupted Bcl-2/Bak heterodimers, allowing Bak to execute the cell death program. Treatment with a Bcl-2-specific BH3 mimetic, ABT-199, reduced tumor formation and tumor burden in a murine model of cholangiocarcinoma. Collectively, these findings indicate that apoptotic priming of CAF by PDGF occurs via Puma-mediated Bak activation, which can be converted to active full-blown apoptosis by navitoclax or ABT-199 for therapeutic benefit.
Collapse
Affiliation(s)
- Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Joachim C Mertens
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich 8091, Switzerland, and
| | - Steven F Bronk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Haiming Dai
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Scott H Kaufmann
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905,.
| |
Collapse
|
7
|
Blazek M, Betz C, Hall MN, Reth M, Zengerle R, Meier M. Proximity ligation assay for high-content profiling of cell signaling pathways on a microfluidic chip. Mol Cell Proteomics 2013; 12:3898-907. [PMID: 24072685 DOI: 10.1074/mcp.m113.032821] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Here, we present the full integration of a proximity ligation assay (PLA) on a microfluidic chip for systematic cell signaling studies. PLA is an in situ technology for the detection of protein interaction, post-translational modification, concentration, and cellular location with single-molecule resolution. Analytical performance advances on chip are achieved, including full automation of the biochemical PLA steps, target multiplexing, and reduction of antibody consumption by 2 orders of magnitude relative to standard procedures. In combination with a microfluidic cell-culturing platform, this technology allows one to gain control over 128 cell culture microenvironments. We demonstrate the use of the combined cell culture and protein analytic assay on chip by characterizing the Akt signaling pathway upon PDGF stimulation. Signal transduction is detected by monitoring the phosphorylation states of Akt, GSK-3β, p70S6K, S6, Erk1/2, and mTOR and the cellular location of FoxO3a in parallel with the PLA. Single-cell PLA results revealed for Akt and direct targets of Akt a maximum activation time of 4 to 8 min upon PDGF stimulation. Activation times for phosphorylation events downward in the Akt signaling pathway including the phosphorylation of S6, p70S6K, and mTOR are delayed by 8 to 10 min or exhibit a response time of at least 1 h. Quantitative confirmation of the Akt phosphorylation signal was determined with the help of a mouse embryonic fibroblast cell line deficient for rictor. In sum, this work with a miniaturized PLA chip establishes a biotechnological tool for general cell signaling studies and their dynamics relevant for a broad range of biological inquiry.
Collapse
Affiliation(s)
- Matthias Blazek
- Microfluidic and Biological Engineering, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
8
|
Bridges RS, Kass D, Loh K, Glackin C, Borczuk AC, Greenberg S. Gene expression profiling of pulmonary fibrosis identifies Twist1 as an antiapoptotic molecular "rectifier" of growth factor signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:2351-61. [PMID: 19893041 DOI: 10.2353/ajpath.2009.080954] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and typically fatal lung disease. To gain insight into IPF pathogenesis, we performed gene expression profiling of IPF lungs. Twist1, a basic helix-loop-helix protein, was found among the most consistently and highly up-regulated genes and was expressed in nuclei of type II epithelial cells, macrophages, and fibroblasts in IPF lungs. We studied the function of Twist1 in fibroblasts further, because they are the major effector cells in this disease and persist despite an ambient proapoptotic environment. Twist1 was induced by the profibrotic growth factors (GFs) basic fibroblast growth factor, platelet-derived growth factor, and epidermal growth factor in primary rat lung fibroblasts (RLFs). Suppression of Twist1 expression resulted in decreased RLF accumulation due to increased apoptosis, whereas Twist1 overexpression protected RLFs against several apoptotic stimuli. Addition of platelet-derived growth factor in combination with other GFs led to an increase in proliferation. When Twist1 was depleted, GFs continued to act as mitogens but caused a marked increase in cell death. The increase in apoptosis under basal or growth factor-stimulated conditions was partly mediated by up-regulation of the proapoptotic Bcl-2 family members, Bim and PUMA. These findings indicate that Twist1 promotes survival and accumulation of fibroblasts by shaping their responsiveness to growth factor stimulation. We propose that Twist1 represents one of the factors that promotes pathogenic accumulation of fibroblasts in fibrotic lung disease.
Collapse
Affiliation(s)
- Robert S Bridges
- Department of Pharmacology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
9
|
Faraone D, Aguzzi MS, Toietta G, Facchiano AM, Facchiano F, Magenta A, Martelli F, Truffa S, Cesareo E, Ribatti D, Capogrossi MC, Facchiano A. Platelet-derived growth factor-receptor alpha strongly inhibits melanoma growth in vitro and in vivo. Neoplasia 2009; 11:732-742. [PMID: 19649203 PMCID: PMC2713586 DOI: 10.1593/neo.09408] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Revised: 04/16/2009] [Accepted: 04/20/2009] [Indexed: 02/08/2023]
Abstract
Cutaneous melanoma is the most aggressive skin cancer; it is highly metastatic and responds poorly to current therapies. The expression of platelet-derived growth factor receptors (PDGF-Rs) is reported to be reduced in metastatic melanoma compared with benign nevi or normal skin; we then hypothesized that PDGF-Ralpha may control growth of melanoma cells. We show here that melanoma cells overexpressing PDGF-Ralpha respond to serum with a significantly lower proliferation compared with that of controls. Apoptosis, cell cycle arrest, pRb dephosphorylation, and DNA synthesis inhibition were also observed in cells overexpressing PDGF-Ralpha. Proliferation was rescued by PDGF-Ralpha inhibitors, allowing to exclude nonspecific toxic effects and indicating that PDGF-Ralpha mediates autocrine antiproliferation signals in melanoma cells. Accordingly, PDGF-Ralpha was found to mediate staurosporine cytotoxicity. A protein array-based analysis of the mitogen-activated protein kinase pathway revealed that melanoma cells overexpressing PDGF-Ralpha show a strong reduction of c-Jun phosphorylated in serine 63 and of protein phosphatase 2A/Balpha and a marked increase of p38gamma, mitogen-activated protein kinase kinase 3, and signal regulatory protein alpha1 protein expression. In a mouse model of primary melanoma growth, infection with the Ad-vector overexpressing PDGF-Ralpha reached a significant 70% inhibition of primary melanoma growth (P < .001) and a similar inhibition of tumor angiogenesis. All together, these data demonstrate that PDGF-Ralpha strongly impairs melanoma growth likely through autocrine mechanisms and indicate a novel endogenous mechanism involved in melanoma control.
Collapse
Affiliation(s)
- Debora Faraone
- Laboratorio Biologia Vascolare e Terapia Genica, Centro Cardiologico Monzino, IRCCS, Milano, Italy
| | - Maria Simona Aguzzi
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| | - Gabriele Toietta
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| | - Angelo M Facchiano
- Laboratorio Bioinformatica e Biologia Computazionale, Istituto di Scienza dell'Alimentazione CNR, Avellino, Italy
| | - Francesco Facchiano
- Dipartimento Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Roma, Italy
| | - Alessandra Magenta
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| | - Fabio Martelli
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| | - Silvia Truffa
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| | - Eleonora Cesareo
- Laboratorio Ingegneria Tessutale e Fisiopatologia Cutanea, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| | | | - Maurizio C Capogrossi
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| | - Antonio Facchiano
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Roma, Italy
| |
Collapse
|
10
|
Stintzing S, Ocker M, Hartner A, Amann K, Barbera L, Neureiter D. Differentiation patterning of vascular smooth muscle cells (VSMC) in atherosclerosis. Virchows Arch 2009; 455:171-185. [PMID: 19557430 DOI: 10.1007/s00428-009-0800-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 05/11/2009] [Accepted: 06/06/2009] [Indexed: 01/17/2023]
Abstract
To investigate the involvement of transdifferentiation and dedifferentiation phenomena inside atherosclerotic plaques, we analyzed the differentiation status of vascular smooth muscle cells (VSMC) in vitro and in vivo. Forty normal autoptic and 20 atherosclerotic carotid endarterectomy specimens as well as 20 specimens of infrarenal and suprarenal aortae were analyzed for the expression of cytokeratins 7 and 18 and beta-catenin as markers (epithelial transdifferentiation) as well as CD31 and CD34 (embryonic dedifferentiation) by conventional and double fluorescence immunohistochemistry and reverse transcription polymerase chain reaction. Looking at these markers, additional cell culture experiments with human aortic (HA)-VSMC were done under stimulation with IL-1beta, IL-6, and TNF-alpha. Cytokeratins and beta-catenin were expressed significantly higher in atherosclerotic than in normal carotids primarily localized in VSMC of the shoulder/cap region of atherosclerotic lesions. Additionally, heterogeneous cellular coexpression of CD31 and/or CD34 was observed in subregions of progressive atherosclerotic lesions by VSMC. The expression of those differentiation markers by stimulated HA-VSMC showed a time and cytokine dependency in vitro. Our findings show that (1) VSMC of progressive atheromas have the ability of differentiation, (2) that transdifferentiation and dedifferentiation phenomena are topographically diverse localized in the subregions of advanced atherosclerotic lesions, and (3) are influenced by inflammatory cytokines like IL-1beta, IL-6, and TNF-alpha.
Collapse
Affiliation(s)
- Sebastian Stintzing
- Department of Medicine III, University Hospital Grosshadern, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
11
|
Harks EGA, Peters PHJ, van Dongen JLJ, van Zoelen EJJ, Theuvenet APR. Autocrine production of prostaglandin F2αenhances phenotypic transformation of normal rat kidney fibroblasts. Am J Physiol Cell Physiol 2005; 289:C130-7. [PMID: 15758043 DOI: 10.1152/ajpcell.00416.2004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have used normal rat kidney (NRK) fibroblasts as an in vitro model system to study cell transformation. These cells obtain a transformed phenotype upon stimulation with growth-modulating factors such as retinoic acid (RA) or transforming growth factor-β (TGF-β). Patch-clamp experiments showed that transformation is paralleled by a profound membrane depolarization from around −70 to −20 mV. This depolarization is caused by a compound in the medium conditioned by transformed NRK cells, which enhances intracellular Ca2+levels and thereby activates Ca2+-dependent Cl−channels. This compound was identified as prostaglandin F2α(PGF2α) using electrospray ionization mass spectrometry. The active concentration in the medium conditioned by transformed NRK cells as determined using an enzyme immunoassay was 19.7 ± 2.5 nM ( n = 6), compared with 1.5 ± 0.1 nM ( n = 3) conditioned by nontransformed NRK cells. Externally added PGF2αwas able to trigger NRK cells that had grown to density arrest to restart their proliferation. This proliferation was inhibited when the FP receptor (i.e., natural receptor for PGF2α) was blocked by AL-8810. RA-induced phenotypic transformation of NRK cells was partially (∼25%) suppressed by AL-8810. Our results demonstrate that PGF2αacts as an autocrine enhancer and paracrine inducer of cell transformation and suggest that it may play a crucial role in carcinogenesis in general.
Collapse
Affiliation(s)
- E G A Harks
- Department of Cell Biology, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
12
|
Lehti K, Allen E, Birkedal-Hansen H, Holmbeck K, Miyake Y, Chun TH, Weiss SJ. An MT1-MMP-PDGF receptor-beta axis regulates mural cell investment of the microvasculature. Genes Dev 2005; 19:979-91. [PMID: 15805464 PMCID: PMC1080136 DOI: 10.1101/gad.1294605] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Platelet-derived growth factor (PDGF)/PDGFRbeta-dependent investment of the vascular endothelium by mural cells (i.e., pericytes and vascular smooth muscle cells; VSMCs) is critical for normal vessel wall structure and function. In the developing vasculature, mural cell recruitment is associated with the functionally undefined expression of the type I transmembrane proteinase, membrane-type 1 matrix metalloproteinase (MT1-MMP). In this paper, using VSMCs and tissues isolated from gene-targeted mice, we identify MT1-MMP as a PDGF-B-selective regulator of PDGFRbeta-dependent signal transduction and mural cell function. In VSMCs, catalytically active MT1-MMP associates with PDGFRbeta in membrane complexes that support the efficient induction of mitogenic signaling by PDGF-B in a matrix metalloproteinase inhibitor-sensitive fashion. In contrast, MT1-MMP-deficient VSMCs display PDGF-B-selective defects in chemotaxis and proliferation as well as ERK1/2 and Akt activation that can be rescued in tandem fashion following retroviral transduction with the wild-type protease. Consistent with these in vitro findings, MT1-MMP-deficient brain tissues display a marked reduction in mural cell density as well as abnormal vessel wall morphology similar to that reported in mice expressing PDGF-B or PDGFRbeta hypomorphic alleles. Together, these data identify MT1-MMP as a novel proteolytic modifier of PDGF-B/PDGFRbeta signal transduction that cooperatively regulates vessel wall architecture in vivo.
Collapse
Affiliation(s)
- Kaisa Lehti
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Wang XZ, Zhang SJ, Chen YX, Chen ZX, Huang YH, Zhang LJ. Effects of platelet-derived growth factor and interleukin-10 on Fas/Fas-ligand and Bcl-2/Bax mRNA expression in rat hepatic stellate cells in vitro. World J Gastroenterol 2004; 10:2706-10. [PMID: 15309723 PMCID: PMC4572197 DOI: 10.3748/wjg.v10.i18.2706] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To investigate the effects of platelet-derived growth factor (PDGF) and interleukin-10 (IL-10) on Fas/Fas-ligand and Bcl-2/Bax mRNA expressions in rat hepatic stellate cells.
METHODS: Rat hepatic stellate cells (HSCs) were isolated and purified from rat liver by in situ digestion of collagenase and pronase and single-step density Nycodenz gradient. After activated by culture in vitro, HSCs were divided into 4 groups and treated with nothing (group N), PDGF (group P), IL-10 (group I) and PDGF in combination with IL-10 (group C), respectively. Semi-quantitative reverse-transcriptase polymerase chain reaction (RT-PCR) analysis was employed to compare the mRNA expression levels of Fas/FasL and Bcl-2/Bax in HSCs of each group.
RESULTS: The expression levels of Fas between the 4 groups had no significant differences (P > 0.05). FasL mRNA level in normal culture-activated HSCs (group N) was very low. It increased obviously after HSCs were treated with IL-10 (group I) (0.091 ± 0.007 vs 0.385 ± 0.051, P < 0.01), but remained the low level after treated with PDGF alone (group P) or PDGF in combination with IL-10 (group C). Contrast to the control group, after treated with PDGF and IL-10, either alone or in combination, Bcl-2 mRNA expression was down-regulated and Bax mRNA expression was up-regulated, both following the turn from group P, group I to group C. Expression of Bcl-2 mRNA in group C was significantly lower than that in group P (0.126 ± 0.008 vs 0.210 ± 0.024, P < 0.01). But no significant difference was found between group C and group I, as well as between group I and group P (P > 0.05). Similarly, the expression of Bax in group C was higher than that in group P (0.513 ± 0.016 vs 0.400 ± 0.022, P < 0.01). No significant difference was found between group I and group P (P > 0.05). But compared with group C, Bax expressions in group I tended to decrease (0.449 ± 0.028 vs 0.513 ± 0.016, P < 0.05).
CONCLUSION: PDGF may promote proliferation of HSCs but is neutral with respect to HSC apoptosis. IL-10 may promote the apoptosis of HSCs by up-regulating the expressions of FasL and Bax and down-regulating the expression of Bcl-2, which may be involved in its antifibrosis mechanism.
Collapse
Affiliation(s)
- Xiao-Zhong Wang
- Department of Gastroenterology, Union Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China.
| | | | | | | | | | | |
Collapse
|
14
|
Tokunou T, Shibata R, Kai H, Ichiki T, Morisaki T, Fukuyama K, Ono H, Iino N, Masuda S, Shimokawa H, Egashira K, Imaizumi T, Takeshita A. Apoptosis induced by inhibition of cyclic AMP response element-binding protein in vascular smooth muscle cells. Circulation 2003; 108:1246-52. [PMID: 12939230 DOI: 10.1161/01.cir.0000085164.13439.89] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The balance between apoptosis and proliferation of vascular smooth muscle cells (VSMCs) is believed to contribute to the vascular remodeling process. Cyclic AMP response element-binding protein (CREB) is a critical transcription factor for the survival of neuronal cells and T lymphocytes. However, the role of CREB in blood vessels is incompletely characterized. METHODS AND RESULTS Nuclear staining with Hoechst 33258 or propidium iodine showed an increase in apoptotic cells with activation of caspase-3 in VSMCs infected with adenovirus expressing the dominant-negative form of CREB (AdCREBM1). Basal expression of Bcl-2 and Bcl-2 promoter activity were decreased by infection with AdCREBM1. Immunohistochemistry revealed that CREB was mainly induced and activated in the neointimal alpha-smooth muscle actin-positive cells of rat carotid artery after balloon injury. Infection with AdCREBM1 suppressed neointimal formation (intima-media ratio) by 33.8% after 14 days of injury, which was accompanied by an increase in apoptosis as indicated by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling-positive cells and a decrease in bromodeoxyuridine incorporation. CONCLUSIONS These results suggest that CRE-dependent gene transcription might play an important role in the survival and proliferation of VSMCs. CREB might be a novel transcription factor mediating the vascular remodeling process and a potential therapeutic target for atherosclerotic disease.
Collapse
MESH Headings
- Angioplasty, Balloon
- Animals
- Apoptosis/drug effects
- Apoptosis/physiology
- Bromodeoxyuridine/pharmacokinetics
- Carotid Artery, Common/drug effects
- Carotid Artery, Common/metabolism
- Cell Division
- Cells, Cultured
- Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors
- Cyclic AMP Response Element-Binding Protein/genetics
- Cyclic AMP Response Element-Binding Protein/metabolism
- Disease Models, Animal
- Gene Expression/drug effects
- Genes, Dominant
- Genetic Vectors/administration & dosage
- Genetic Vectors/genetics
- In Situ Nick-End Labeling
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Rats
- Transfection
- Tunica Intima/drug effects
- Tunica Intima/metabolism
Collapse
Affiliation(s)
- Tomotake Tokunou
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, 812-8582 Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Buetow BS, Tappan KA, Crosby JR, Seifert RA, Bowen-Pope DF. Chimera analysis supports a predominant role of PDGFRbeta in promoting smooth-muscle cell chemotaxis after arterial injury. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:979-84. [PMID: 12937138 PMCID: PMC1868260 DOI: 10.1016/s0002-9440(10)63457-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The carotid artery shows a common response to many forms of injury, including a rapid activation of smooth muscle cell (SMC) proliferation in the media and migration of SMCs into the intima to form a neointima. Platelet-derived growth factor (PDGF) is believed to play a role in this response to injury, but it has proven difficult to distinguish whether it is stimulating cell migration or cell proliferation, and whether the action is direct or indirect. To determine this, we created chimeric mice composed of both wild-type (WT) and marked PDGF receptor beta (PDGFRbeta)-deficient cells, and determined the consequences of PDGFRbeta expression for SMC participation in response to ligation of the left common carotid artery. The proportion of PDGFRbeta-/- SMCs increased 4.5-fold in the media and decreased 1.8-fold during formation of the neointima, consistent with migration of WT SMCs out of the media and into the intima, leaving the PDGFRbeta-/- cells behind. The fibrotic reaction in the adventitia, which does not involve cell migration, did not result in any change in relative abundance of WT and PDGFRbeta-deficient fibroblasts. We conclude that the most significant direct role of PDGFRbeta is to mediate responses that involve cell migration rather than proliferation.
Collapse
Affiliation(s)
- Bernard S Buetow
- Department of Pathology, University of Washington, Seattle, WA 98195-7470, USA
| | | | | | | | | |
Collapse
|
16
|
Anto RJ, Venkatraman M, Karunagaran D. Inhibition of NF-kappaB sensitizes A431 cells to epidermal growth factor-induced apoptosis, whereas its activation by ectopic expression of RelA confers resistance. J Biol Chem 2003; 278:25490-8. [PMID: 12714587 DOI: 10.1074/jbc.m301790200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epidermal growth factor (EGF) is a well known mitogen, but it paradoxically induces apoptosis in cells that overexpress its receptor. We demonstrate for the first time that the EGF-induced apoptosis is accelerated if NF-kappaB is inactivated. To inactivate NF-kappaB, human epidermoid carcinoma cells (A431) that overexpress EGF receptor were stably transfected with an IkappaB-alpha double mutant construct. Under the NF-kappaB-inactivated condition, A431 cells were more sensitive to EGF with decreased cell viability and increased externalization of phosphatidylserine on the cell surface, DNA fragmentation, and activation of caspases (3 and 8 but not 9), typical features of apoptosis. These results were further supported by the potentiation of the growth inhibitory effects of EGF by chemical inhibitors of NF-kappaB (curcumin and sodium salicylate) and the protective role of RelA evidenced by the resistance of A431-RelA cells (stably transfected with RelA) to EGF-induced apoptosis. EGF treatment or ectopic expression of RelA in A431 cells induced DNA binding activity of NF-kappaB (p50 and RelA) and the expression of c-IAP1, a downstream target of NF-kappaB. A431-RelA cells exhibited spontaneous phosphorylation of Akt (a downstream target of phosphatidylinositol 3-kinase and regulator of NF-kappaB) and EGF treatment stimulated it further. Blocking this basal Akt phosphorylation with LY294002, an inhibitor of phosphatidylinositol 3-kinase, did not affect their viability but blocking of EGF-induced phosphorylation of Akt sensitized the otherwise resistant A431-RelA cells to EGF-mediated growth inhibition. Our results favor an anti-apoptotic role for NF-kappaB in the regulation of EGF-induced apoptosis.
Collapse
Affiliation(s)
- Ruby John Anto
- Division of Cancer Biology, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, Kerala-695014, India
| | | | | |
Collapse
|
17
|
Miyashita Y, Ozaki H, Koide N, Otsuka M, Oyama T, Itoh Y, Mastuzaka T, Shirai K. Oxysterol-induced apoptosis of vascular smooth muscle cells is reduced by HMG-CoA reductase inhibitor, pravastatin. J Atheroscler Thromb 2003; 9:65-71. [PMID: 12238640 DOI: 10.5551/jat.9.65] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
We investigated the mechanism by which 7-ketocholesterol damages vascular smooth muscle cells and the protective effect of the hydroxymethyl glutary CoA reductase inhibitor, pravastatin on it. When 7-ketocholesterol (50 micromol/L) was added to cultured human vascular smooth muscle cells, the extent of cell detachment increased and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling was positive. DNA extracted from the smooth muscle cells exposed to 7-ketocholesterol showed a ladder pattern on agarose electrophoresis. The fragmented DNA also increased in smooth muscle cells incubated with 7-ketocholesterol dose-dependently. In the presence of pravastatin, the cell detachment induced by 7-ketocholesterol was inhibited and the amount of fragmented DNA decreased significantly. These effects of pravastatin were inhibited by mevalonate. The results suggest that 7-ketocholesterol-induced apoptosis of vascular smooth muscle cells is inhibited by pravastatin, and mevalonate acts as a trigger of the apoptosis.
Collapse
Affiliation(s)
- Yoh Miyashita
- Center of Diabetes, Endocrinology & Metabolism, Sakura Hospital, Toho University School of Medicine, Chiba, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Yu J, Ustach C, Kim HRC. Platelet-derived growth factor signaling and human cancer. JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2003; 36:49-59. [PMID: 12542975 DOI: 10.5483/bmbrep.2003.36.1.049] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Platelet-derived growth factor (PDGF) is a critical regulator of mesenchymal cell migration and proliferation. The vital functions of PDGFs for angiogenesis, as well as development of kidney, brain, cardiovascular system and pulmonary alveoli during embryogenesis, have been well demonstrated by gene knock-out approaches. Clinical studies reveal that aberrant expression of PDGF and its receptor is often associated with a variety of disorders including atherosclerosis, fibroproliferative diseases of lungs, kidneys and joints, and neoplasia. PDGF contributes to cancer development and progression by both autocrine and paracrine signaling mechanisms. In this review article, important features of the PDGF isoforms and their cell surface receptor subunits are discussed, with regards to signal transduction, PDGF-isoform specific cellular responses, and involvement in angiogensis, and tumorstromal interactions.
Collapse
Affiliation(s)
- Jiuhong Yu
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
19
|
Mitsuda T, Furukawa K, Fukumoto S, Miyazaki H, Urano T, Furukawa K. Overexpression of ganglioside GM1 results in the dispersion of platelet-derived growth factor receptor from glycolipid-enriched microdomains and in the suppression of cell growth signals. J Biol Chem 2002; 277:11239-46. [PMID: 11782461 DOI: 10.1074/jbc.m107756200] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To investigate the molecular mechanisms of gangliosides for the regulation of cell proliferation, Swiss 3T3 cells were transfected with GM2/GD2 synthase and GM1 synthase cDNAs, resulting in the establishment of GM1-expressing (GM1(+)) lines. Compared with the vector control (GM1(-)) cell lines, GM1(+) cells exhibited reduced cell proliferation by stimulation with platelet-derived growth factor (PDGF). In accordance with the reduced cell growth, GM1(+) cells showed earlier decreases in the phosphorylation levels of PDGF receptor and less activation of MAP kinases than GM1(-) cells. To analyze the effects of GM1 expression on the PDGF/PDGF receptor (PDGFR) signals, the glycolipid-enriched microdomain (GEM) was isolated and the following results were obtained. (i) PDGFR predominantly distributed in the non-GEM fraction in GM1(+) cells, while it was present in both GEM and non-GEM fractions in GM1(-) cells. (ii) Activation of PDGFR as detected by anti-phosphotyrosine antibody occurred almost in parallel with existing amounts of PDGFR in each fraction. (iii) GM1 binds with PDGFR in GEM fractions. These findings suggested that GM1 regulates signals via PDGF/PDGFR by controlling the distribution of PDGFR in- and outside of GEM, and also interacting with PDGFR in the GEM fraction as a functional constituent of the microdomain.
Collapse
Affiliation(s)
- Teruhiko Mitsuda
- Department of Biochemistry II, Nagoya University School of Medicine, 65 Tsurumai, Nagoya, 466-0065 Japan
| | | | | | | | | | | |
Collapse
|
20
|
Deb A, Zamanian-Daryoush M, Xu Z, Kadereit S, Williams BR. Protein kinase PKR is required for platelet-derived growth factor signaling of c-fos gene expression via Erks and Stat3. EMBO J 2001; 20:2487-96. [PMID: 11350938 PMCID: PMC125453 DOI: 10.1093/emboj/20.10.2487] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The double-stranded RNA (dsRNA)-activated protein kinase PKR is an interferon (IFN)-induced enzyme that controls protein synthesis through phosphorylation of eukaryotic initiation factor 2alpha (eIF-2alpha). PKR also regulates signals initiated by diverse stimuli, including dsRNA, IFN-gamma, tumor necrosis factor-alpha, interleukin-1 and lipopolysaccharide, to different transcription factors, resulting in pro-inflammatory gene expression. Stat3 plays an essential role in promoting cell survival and proliferation by different growth factors, including platelet-derived growth factor (PDGF). Here we show that PKR physically interacts with Stat3 and is required for PDGF-induced phosphorylation of Stat3 at Tyr705 and Ser727, resulting in DNA binding and transcriptional activation. PKR-mediated phosphorylation of Stat3 on Ser727 is indirect and channeled through ERKS: Although PKR is pre-associated with the PDGF beta-receptor, treatment with PDGF only modestly activates PKR. However, the induction of c-fos by PDGF is defective in PKR-null cells. Taken together, these results establish PKR as an upstream regulator of activation of Stat3 and as a common mediator of both growth-promoting and growth-inhibitory signals.
Collapse
Affiliation(s)
| | | | | | | | - Bryan R.G. Williams
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
Corresponding author e-mail:
| |
Collapse
|
21
|
Metheny-Barlow LJ, Flynn B, van Gijssel HE, Marrogi A, Gerwin BI. Paradoxical effects of platelet-derived growth factor-A overexpression in malignant mesothelioma. Antiproliferative effects in vitro and tumorigenic stimulation in vivo. Am J Respir Cell Mol Biol 2001; 24:694-702. [PMID: 11415934 DOI: 10.1165/ajrcmb.24.6.4334] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Malignant mesothelioma is associated with asbestos exposure and remains resistant to all therapeutic intervention. Previous studies have suggested an enhancing role for platelet-derived growth factor (PDGF) in mesothelial tumorigenicity, although the mechanism by which PDGF facilitates tumorigenicity is unknown. Here, we evaluate the contribution of PDGF-A expression to mesothelial tumorigenicity using ectopic modulation of PDGF-A expression. We find, in accordance with other reports, that the receptor for PDGF-A, although expressed at high levels in normal human mesothelial cells, is not easily detectable in mesothelioma. Further, we show that PDGF-A overexpression is responsible for autocrine downregulation of its receptor. Our data indicate, surprisingly, that for mesothelioma cells in vitro, high-level activation of a PDGF-A-PDGF receptor loop is antiproliferative whereas abrogation of PDGF-A expression stimulates growth. These data suggest that PDGF-A does not contribute to tumorigenicity by autocrine stimulation of proliferation. In contrast, increased PDGF-A expression in vivo increases tumor incidence and growth rate and decreases the latency period to tumor formation whereas abrogation of PDGF-A expression decreases tumor incidence and increases latency. Thus, the tumorigenic effect of PDGF-A must act through paracrine mechanisms relevant at early stages of tumor initiation.
Collapse
Affiliation(s)
- L J Metheny-Barlow
- Laboratory of Human Carcinogenesis; and Laboratory of Cellular Carcinogenesis and Tumor Promotion, Division of Basic Sciences, National Cancer Institute, Bethesda, Maryland, USA.
| | | | | | | | | |
Collapse
|
22
|
Yu J, Moon A, Kim HR. Both platelet-derived growth factor receptor (PDGFR)-alpha and PDGFR-beta promote murine fibroblast cell migration. Biochem Biophys Res Commun 2001; 282:697-700. [PMID: 11401517 DOI: 10.1006/bbrc.2001.4622] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell motility plays a critical role for many physiological and pathological processes including wound healing, fibrosis, angiogenesis, and tumor metastasis. Platelet-derived growth factor (PDGF) is among the most potent stimuli for mesenchymal cell migration. The PDGF B-chain homodimer PDGF BB activates both alpha- and beta-receptor subunits (alpha-PDGFR and beta-PDGFR), and promotes cell migration in many cell types including fibroblasts and smooth muscle cells. PDGF-A chain homodimer PDGF AA activates alpha-PDGFR only, and its role for cell migration is still debatable. PDGF BB, but not PDGF AA, induces smooth muscle cell migration. Interestingly, alpha-PDGFR was shown to antagonize beta-PDGFR-induced smooth muscle cell migration. In the present study, we investigated the role of alpha-PDGFR and beta-PDGFR in PDGF-mediated cell migration of murine fibroblasts (NIH 3T3). Unlike smooth muscle cells, both PDGF AA and PDGF BB promoted NIH 3T3 cell migration. The effect of PDGF BB activation of beta-PDGFR alone for cell migration was examined using previously established NIH 3T3 clones in which alpha-PDGFR signaling is inhibited by a dominant-negative alpha-PDGFR, or an antisense construct of alpha-PDGFR. PDGF BB activation of beta-PDGFR alone was sufficient to induce cell migration, but the efficiency was significantly lower compared to PDGF activation of both receptors. These results showed that both alpha- and beta-PDGFRs promote fibroblast cell migration and their effects are additive. Taken together, we propose that cell-type specific alpha-PDGFR signaling is critical for regulation of mesenchymal cell migration in response to PDGF isoform, whereas beta-PDGFR mainly promotes cell migration.
Collapse
Affiliation(s)
- J Yu
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
23
|
Issa R, Williams E, Trim N, Kendall T, Arthur MJ, Reichen J, Benyon RC, Iredale JP. Apoptosis of hepatic stellate cells: involvement in resolution of biliary fibrosis and regulation by soluble growth factors. Gut 2001; 48:548-57. [PMID: 11247901 PMCID: PMC1728234 DOI: 10.1136/gut.48.4.548] [Citation(s) in RCA: 245] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Activated hepatic stellate cells (HSC) are central to the pathogenesis of liver fibrosis, both as a source of fibrillar collagens that characterise fibrosis and matrix degrading metalloproteinases and their tissue inhibitors, the TIMPs. AIMS To test the hypothesis that HSC apoptosis is critical to recovery from biliary fibrosis and that soluble growth factors may regulate HSC survival and apoptosis. METHODS Rats (n=15) were subjected to bile duct ligation for 21 days, after which biliodigestive anastomosis was undertaken (n=13). Livers were harvested at fixed time points of recovery for periods of up to 42 days. Numbers of activated HSCs were quantified after alpha smooth muscle actin staining and HSC apoptosis was detected by terminal UDP-nick end labelling (TUNEL) staining and quantified at each time point. HSC apoptosis was quantified in vitro in the presence or absence of insulin-like growth factor (IGF)-1, IGF-2, platelet derived growth factor (PDGF), and transforming growth factor beta1 (TGF-beta1). RESULTS Following biliodigestive anastomosis after 21 days of bile duct ligation, rat liver demonstrated a progressive resolution of biliary fibrosis over 42 days, associated with a fivefold decrease in activated HSC determined by alpha smooth muscle actin staining. TUNEL staining indicated that loss of activated HSC resulted from an increase in the rate of apoptosis during the first two days post biliodigestive anastomosis. Serum deprivation and culture in the presence of 50 microM cycloheximide was associated with an increase in HSC apoptosis which was significantly inhibited by addition of 10 ng/ml and 100 ng/ml IGF-1, respectively (0.05>p, n=5). In contrast, 1 and 10 ng/ml of TGF-beta1 caused a significant increase in HSC apoptosis compared with serum free controls (p<0.05, n=4). PDGF and IGF-2 were neutral with respect to their effect on HSC apoptosis. CONCLUSION HSC apoptosis plays a critical role in the spontaneous recovery from biliary fibrosis. Both survival and apoptosis of HSC are regulated by growth factors expressed during fibrotic liver injury.
Collapse
Affiliation(s)
- R Issa
- Liver Research Group, Division of Cell and Molecular Medicine, Level D, South Lab and Path Block, Southampton General Hospital, Southampton, UK
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Apoptosis is a cell suicide program characterized by distinct morphological (cell shrinkage, membrane blebbing, pyknosis, chromatin margination, denser cytoplasmic images) and biochemical (e.g., DNA fragmentation into distinct ladders; degradation of apoptotic markers such as PARP and nuclear lamins) features. It is involved in multiple physiological processes examplified by involution of mammary tissues, embryonic development, homeostatic maintenance of tissues and organs, and maturation of the immune system, as well as in many pathological conditions represented by neurologic degeneration (Alzeimer's disease), autoimmune and inflammatory diseases, etiology of atherosclerosis, AIDS, and oncogenesis and tumor progression. Numerous molecular entities have been shown to regulate the apoptotic process. This review provides a concise summary of the recent data on the role of oncogenes/tumor suppressor genes, cytokines and growth factors/growth factor receptors, intracellular signal transducers, cell cycle regulators, reactive oxygen species or other free radicals, extracellular matrix regulators/cell adhesion molecules, and specific endonucleases and cytoplasmic proteases (the ICE family proteins) in regulating cell survival and apoptosis. Elucidation of the molecular mechanisms regulating apoptosis bears tremendous impact on enhancing our understanding of many diseases inflicting the human beings and undoubtedly brings us hope for the cure of these diseases.
Collapse
Affiliation(s)
- Dean G Tang
- Wayne State University, Department of Radiation Oncology, Detroit, USA
| | | |
Collapse
|
25
|
Bruserud O, Foss B, Abrahamsen JF, Gjertsen BT, Ernst P. Autologous stem cell transplantation as post-remission therapy in adult acute myelogenous leukemia: does platelet contamination of peripheral blood mobilized stem cell grafts influence the risk of leukemia relapse? JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2000; 9:433-43. [PMID: 10982241 DOI: 10.1089/152581600419099] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Conventional chemotherapy of acute myelogenous leukemia (AML) results in an overall long-term disease-free survival of less than 50%, but for selected subsets of younger patients the prognosis can be improved by allogeneic stem cell transplantation. The use of autologous stem cell transplantation is now investigated as an alternative to allotransplantation due to its lower risk of serious complications. However, autotransplantation is associated with a relatively high risk of post-transplant AML relapse that can be derived from contaminating leukemia cells in the autograft. Peripheral blood mobilized stem cell (PBSC) grafts usually contain a higher number of platelets. The degree of platelet contamination is determined by the peripheral blood platelet count at the time of harvesting, and the platelets become activated and release soluble mediators during the ex vivo handling of PBSC grafts. Many of these platelet-derived mediators can bind to specific receptors expressed by AML blasts, and the platelet contamination may then alter AML blast survival and thereby influence the risk of post-transplant leukemia relapse. Therefore, we conclude that the platelet contamination of autologous stem cell grafts is possibly of clinical importance, but the effect of this nonstandardized parameter is difficult to predict in individual patients because the number of graft-contaminating platelets, the degree of platelet activation, and the effects of platelet-derived mediators on AML blasts differ between patients.
Collapse
Affiliation(s)
- O Bruserud
- Department of Medicine, Haukeland University Hospital and the University of Bergen, Norway
| | | | | | | | | |
Collapse
|
26
|
Yu J, Deuel TF, Kim HR. Platelet-derived growth factor (PDGF) receptor-alpha activates c-Jun NH2-terminal kinase-1 and antagonizes PDGF receptor-beta -induced phenotypic transformation. J Biol Chem 2000; 275:19076-82. [PMID: 10777515 DOI: 10.1074/jbc.m910329199] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-derived growth factor (PDGF) is a potent mitogen for mesenchymal cells. The PDGF B-chain (c-sis proto-oncogene) homodimer (PDGF BB) and v-sis, its viral counterpart, activate both alpha- and beta-receptor subunits (alpha-PDGFR and beta-PDGFR) and mediate anchorage-independent growth in NIH3T3 cells. In contrast, the PDGF A chain homodimer (PDGF AA) activates alpha-PDGFR only and fails to induce phenotypic transformation. In the present study, we investigated alpha- and beta-PDGFR specific signaling pathways that are responsible for the differences between the transforming ability of PDGF AA and BB. To study PDGF BB activation of beta-PDGFR, we established NIH3T3 clones in which alpha-PDGFR signaling is inhibited by a dominant-negative alpha-PDGFR, or an antisense construct of alpha-PDGFR. Here, we demonstrate that beta-PDGFR activation alone is sufficient for PDGF BB-mediated anchorage-independent cell growth. More importantly, inhibition of alpha-PDGFR signaling enhanced PDGF BB-mediated phenotypic transformation, suggesting that alpha-PDGFR antagonizes beta-PDGFR-induced transformation. While both alpha- and beta-receptors effectively activate ERKs, alpha-PDGFR, but not beta-PDGFR, activates stress-activated protein kinase-1/c-Jun NH(2)-terminal kinase-1 (JNK-1). Inhibition of JNK-1 activity using a dominant-negative JNK-1 mutant markedly enhanced PDGF BB-mediated anchorage-independent cell growth, demonstrating an antagonistic role for JNK-1 in PDGF-induced transformation. Consistently, overexpression of wild-type JNK-1 reduced PDGF BB-mediated transformation. Taken together, the present study showed that alpha- and beta-PDGFRs differentially regulate Ras-mitogen-activated protein kinase pathways critical for regulation of cell transformation, and transformation suppressing activity of alpha-PDGFR involves JNK-1 activation.
Collapse
Affiliation(s)
- J Yu
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
27
|
Kamer AR, Krebs L, Hoghooghi SA, Liebow C. Proliferative and apoptotic responses in cancers with special reference to oral cancer. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2000; 10:58-78. [PMID: 10759427 DOI: 10.1177/10454411990100010301] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The study of signal transduction pathways for mechanisms of apoptosis and proliferation has significantly advanced our understanding of human cancer, subsequently leading to more effective treatments. Discoveries of growth factors and oncogenes, especially those that function through phosphorylation on tyrosine residues, have greatly benefited our appreciation of the biology of cancer. The regulation of proliferation and apoptosis through phosphorylation via tyrosine kinases and phosphatases is discussed, as well as the contributions of other systems, such as serine and threonine kinases and phosphatases. Receptors with seven-transmembrane domains, steroid hormones, genes, and "death domains" will also be discussed. This review attempts to compare the regulation of the growth of normal tissues and cancers with an effort to highlight the current knowledge of these factors in the growth regulation of oral/oropharyngeal cancers. Despite the strides made in our understanding of growth regulation in human cancers, the study of oral/oropharyngeal cancer specifically lags behind. More research must be done to further our understanding of oral cancer biology, if we are to develop better, more effective treatment protocols.
Collapse
Affiliation(s)
- A R Kamer
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, State University of New York at Buffalo, 14214, USA
| | | | | | | |
Collapse
|
28
|
|
29
|
Fatatis A, Miller RJ. Cell cycle control of PDGF-induced Ca(2+) signaling through modulation of sphingolipid metabolism. FASEB J 1999; 13:1291-301. [PMID: 10428754 DOI: 10.1096/fasebj.13.11.1291] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The effects of growth factors have been shown to depend on the position of a cell in the cell cycle. However, the physiological basis for this phenomenon remains unclear. Here we show that the majority of both CEINGE clone3 (cl3) and human embryonic kidney 293 cells, when arrested in a quiescent phase (G(0)), responded to platelet-derived growth factor BB (PDGF-BB) with non-oscillatory Ca(2+) signals. Furthermore, the same type of Ca(2+) response was also observed in CEINGE cl3 cells (and to a lesser extent in HEK 293 cells) blocked at the G(1)/S boundary. In contrast, CEINGE cl3 cells synchronized in early G(1) or released from G(1)/S arrest responded in an oscillatory fashion. This cell cycle-dependent modulation of Ca(2+) signaling was not observed on epidermal growth factor and G-protein-coupled receptor stimulation and was not due to differences in the expression of PDGF receptors (PDGFRs) during the cell cycle. We demonstrate that inhibition of sphingosine-kinase, which converts sphingosine to sphingosine-1-phosphate, caused G(0) as well as G(1)/S synchronized cells to restore the oscillatory Ca(2+) response to PDGF-BB. In addition, we show that the synthesis of sphingosine and sphingosine-1-phosphate is regulated by the cell cycle and may underlie the differences in Ca(2+) signaling after PDGFR stimulation.
Collapse
Affiliation(s)
- A Fatatis
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
30
|
Jenkins M, Keir M, McCune JM. Fas Is Expressed Early in Human Thymocyte Development But Does Not Transmit an Apoptotic Signal. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.3.1195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
We investigated the expression and function of Fas on human thymocytes prepared from fetal and pediatric tissue specimens and from SCID-hu Thy/Liv grafts. Unlike mouse thymocytes, human thymocytes exhibited a pattern of Fas expression skewed to immature cells, in that the highest expression was seen on double negative thymocytes and on intrathymic T progenitor cells. Fas expression was intermediate on double positive human thymocytes, and low or negative on mature single positive CD4 and CD8 medullary thymocytes. In spite of this relatively abundant surface expression, cross-linking of Fas with agonist mAb was incapable of triggering an apoptotic signal in human thymocytes. Apoptotic signaling was not enhanced by treatment with cycloheximide, nor by restoring a cosignaling milieu by addition of thymic stromal cells. Mouse thymocytes were induced to apoptosis by cross-linked recombinant soluble human Fas ligand both in vitro and in vivo, though human thymocytes were also resistant to this mode of receptor ligation. Membrane-bound Fas ligand also induced apoptotic death in murine thymocytes but not in human thymocytes. Human thymocytes were as sensitive as Jurkat cells, however, to apoptosis induced by TNF-α, suggesting that these cells have a signaling defect before activation of the earliest caspases. These data demonstrate a durable and specific resistance of human thymocytes to apoptosis induced through Fas receptor engagement, and reveal significant species-specific differences in the biology of thymocyte-programmed cell death.
Collapse
Affiliation(s)
| | - Mary Keir
- *Gladstone Institute of Virology and Immunology, and
| | - Joseph M. McCune
- *Gladstone Institute of Virology and Immunology, and
- †Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, CA 94141
| |
Collapse
|
31
|
Abstract
Adequate control of survival or programmed cell death (apoptosis) of cardiovascular cells appears as an important drug target. While prevention of apoptotic death of cardiomyocytes has been assessed in detail, selective induction of apoptosis of vascular smooth muscle cells or fibroblasts could also be of relevance. Thus, induction of apoptosis of vascular smooth muscle cells by p65 NF-kappa B and Bcl-xL antisense oligonucleotides or p53 overexpression could be useful for limiting vascular lesions associated with restenosis. Although fibroblasts represent the majority of cardiac cells, few attempts were made to induce fibroblast apoptosis in disorders associated with excessive collagen deposition and fibrosis. It is hypothesized that early interference with fibroblast proliferation after myocardial infarction or inflammatory heart disease limits fibrosis which further impairs cardiac performance. A candidate approach could involve growth factor analogues which are known to induce fibroblast apoptosis when an incomplete growth stimulus persists.
Collapse
Affiliation(s)
- H Rupp
- Department of Internal Medicine and Cardiology, Philipps-University of Marburg, Germany.
| | | |
Collapse
|
32
|
Kahn NN. Platelet-stimulated thrombin and PDGF are normalized by insulin and Ca2+ channel blockers. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:E856-62. [PMID: 10329979 DOI: 10.1152/ajpendo.1999.276.5.e856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Coronary artery disease is accelerated in chronic spinal cord injury (SCI). Because prostacyclin (PGI2) may retard atherogenesis through its inhibitory effects on platelet function, the role of PGI2 on SCI platelets was determined. The SCI platelets were neither hypersensitive to aggregating agonists nor resistant to the inhibitory effect of PGI2, but PGI2 failed to inhibit platelet-stimulated thrombin generation and the release of platelet-derived growth factor (PDGF) in SCI. Because thrombin and PDGF are atherogenic mitogens, the generation of these mitogens was investigated. Both the release of PDGF and thrombin generation in SCI platelets were higher when compared with control (n = 12). Treatment of non-SCI platelets with 100 nM PGE1 (a stable probe of PGI2) inhibited the release of the mitogens by 90% (P < 0.001), with no effect on SCI platelets. Scatchard analysis of prostaglandin E1 (PGE1) binding showed a 70% decrease of PGI2 receptors on the SCI platelet surface. Treatment of SCI platelets with insulin or Ca2+ channel blockers restored the PGI2-receptor number and "normalized" the inhibition of PDGF release and thrombin generation by PGI2.
Collapse
Affiliation(s)
- N N Kahn
- Department of Medicine, Mount Sinai School of Medicine, New York 10029, USA.
| |
Collapse
|
33
|
Godbout JP, Cengel KA, Cheng SL, Minshall C, Kelley KW, Freund GG. Insulin activates caspase-3 by a phosphatidylinositol 3'-kinase-dependent pathway. Cell Signal 1999; 11:15-23. [PMID: 10206340 DOI: 10.1016/s0898-6568(98)00024-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Activation of the caspase proteases by c-Jun N-terminal kinase 1 (JNK1) has been proposed as a mechanism of apoptotic cell death. Here we report that insulin activates caspase-3 by a pathway requiring phosphatidylinositol 3'-kinase (PI3-kinase). JNK1 assays demonstrated that insulin treatment of myeloma cells induced 3-fold activation of JNK1. Inhibition of PI3-kinase with wortmannin and LY294002 blocked insulin-dependent activation of JNK1. Caspase assays demonstrated that insulin increased caspase-3 activity 3-fold and that inhibition of PI3-kinase blocked this effect. Cell death was doubled by insulin and was due to a 3-fold increase in apoptosis of cells in the G1/G0 phase of the cell cycle. Inhibition of PI3-kinase completely blocked this effect. Finally, inhibition of caspase-3 with benzyloxycarbonyl-Asp-2,6-dichlorobenzoyloxymethylketone blocked cell death due to insulin. Taken together, these findings indicate that insulin activates caspase-3 by a PI3-kinase-dependent pathway resulting in increased apoptosis and cell death.
Collapse
Affiliation(s)
- J P Godbout
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana 61801, USA
| | | | | | | | | | | |
Collapse
|
34
|
Gong W, Pecci A, Roth S, Lahme B, Beato M, Gressner AM. Transformation-dependent susceptibility of rat hepatic stellate cells to apoptosis induced by soluble Fas ligand. Hepatology 1998; 28:492-502. [PMID: 9696016 DOI: 10.1002/hep.510280229] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cytokine-driven activation of hepatic stellate cells (HSC) in tissue injury and inflammation is a key pathogenetic event in liver fibrogenesis leading to an expanded pool of matrix producing myofibroblasts (MFB) which represent the transformed counterpart of HSC. We hypothesize that expansion of the pool of MFB might also be accomplished by modulation of apoptosis, which plays an opposite and complementary role to mitosis in the cellular homeostasis. We characterized the susceptibility of HSC in primary culture and of MFB in secondary culture to apoptosis induced by the soluble Fas ligand (sFasL) and related the effects to the expression levels of Fas (APO-1/CD95) and some major proapoptotic and contra-apoptotic protooncogenes. MFB showed a dose-dependent apoptotic reaction upon exposure to sFasL as evidenced by a strong increase of nucleosomal DNA fragments, loss of cellular DNA, positive TUNEL reaction, and annexin staining. The effect was found only if protein synthesis (cycloheximide) or RNA synthesis (actinomycin D) were arrested. HSC maintained for various times in primary culture were completely resistant to sFasL in combination with cycloheximide, but in late primary cultures (day 7 onward) an increasing susceptibility to sFasL-mediated apoptosis was developed. By semiquantitative reverse-transcriptase polymerase chain reaction (RT-PCR) analysis and alkaline phosphatase-anti-alkaline phosphatase staining Fas receptor was identified both in HSC and MFB at comparable expression levels. The expression of the contra-apoptotic protooncogenes bcl-2 and bcl-xl was found to be much stronger in early HSC than in late HSC and MFB as shown by ribonuclease protection assay. The expression of bcl-2 was additionally confirmed by semiquantitative RT-PCR and immunoblotting. Proapoptotic bax was found in comparable quantities at the RNA level in HSC and MFB but at the protein level MFB showed increased bax expression. It is concluded that transformation of HSC to MFB is paralleled by an increasing sensitivity to sFasL-mediated apoptosis, which might be related to a strong decrease of bcl-2 and bcl-xl expression, leading to a preponderance of proapoptotic gene expression in MFB. Modulation of apoptotic susceptibility of transforming HSC could be an important complementary pathway in the pathogenesis of fibrosis.
Collapse
Affiliation(s)
- W Gong
- Institute of Clinical Chemistry, Philipps-University, Marburg, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Okura T, Igase M, Kitami Y, Fukuoka T, Maguchi M, Kohara K, Hiwada K. Platelet-derived growth factor induces apoptosis in vascular smooth muscle cells: roles of the Bcl-2 family. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1403:245-53. [PMID: 9685664 DOI: 10.1016/s0167-4889(98)00065-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Apoptosis (programmed cell death) is observed in vascular smooth muscle cells (VSMC) in atherosclerotic lesions and stenotic lesions after injury, and modulates the cellularity of these lesions. It is recognized that cell growth and apoptosis are two linked processes. Platelet-derived growth factor (PDGF) induces VSMC proliferation and migration in vitro. We studied the effect of PDGF on apoptosis in VSMC. Cultured rat VSMC were treated with PDGF-AA or PDGF-BB. PDGF-BB induced cell death in cultured VSMC in a time- and dose-dependent manner, but PDGF-AA did not. Gel electrophoresis of genomic DNA and in situ DNA labeling confirmed that the cell death induced by PDGF-BB is apoptosis. PDGF-BB treatment reduced bcl-2 mRNA and bcl-xl mRNA expression, in contrast, induced bcl-xs mRNA expression, linked with the induction of apoptosis in cultured VSMC.
Collapse
Affiliation(s)
- T Okura
- Second Department of Internal Medicine, Ehime University School of Medicine, Onsen-gun, Ehime 791-0295, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
Jelaska A, Korn JH. Anti-Fas induces apoptosis and proliferation in human dermal fibroblasts: differences between foreskin and adult fibroblasts. J Cell Physiol 1998; 175:19-29. [PMID: 9491777 DOI: 10.1002/(sici)1097-4652(199804)175:1<19::aid-jcp3>3.0.co;2-f] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Apoptosis, or programmed cell death, is a naturally occurring process mediated by extracellular signals. We studied anti-Fas (CD95/Apo-1) antibody-induced apoptosis in cultured human foreskin and adult dermal fibroblasts. Induction of apoptosis was identified by fluorescence in situ DNA end-labeling. Anti-Fas antibody induced apoptosis in fibroblasts in a dose- and time-dependent manner. Adult dermal skin fibroblasts were more susceptible to anti-Fas antibody-induced apoptosis than foreskin fibroblasts, with 21-52% dead cells in different strains. In foreskin fibroblasts, anti-Fas antibody (1.0 microg/ml) predominantly induced proliferation ([3H]thymidine incorporation increased to 115-165% of control level) and only low levels of apoptotic cell death after 48 hours of treatment. No induction of proliferation by anti-Fas was found in the adult fibroblasts. Addition of tumor necrosis factor-alpha (TNF-alpha) slightly augmented the anti-Fas antibody-induced apoptosis in both cell types. When we examined the levels of Fas expression using flow cytometry, we found two- to threefold higher Fas expression in adult fibroblasts. C6-ceramide treatment, which induces Fas-independent apoptosis, gave similar levels of cell death in both foreskin and adult fibroblasts. No proliferation was observed in C6-ceramide-treated fibroblasts. Thus, this difference in apoptosis between adult dermal and foreskin fibroblasts appears to be related to the level of Fas expression. When clones of foreskin fibroblasts were examined, there was heterogeneity of anti-Fas antibody-induced apoptosis and proliferation in the cloned fibroblast subpopulations, but this was not correlated with differences in Fas expression. Alterations in fibroblast populations during the process of differentiation and aging may result from selective loss of apoptosis-susceptible populations.
Collapse
Affiliation(s)
- A Jelaska
- Department of Medicine, The Arthritis Center, Boston University School of Medicine, Massachusetts 02118, USA
| | | |
Collapse
|
37
|
Abstract
In eukaryotes, the regulation of tissue cell numbers is a critical homeostatic objective that is achieved through tight control of apoptosis, mitosis and differentiation. While much is known about the genetic regulation of cell growth and differentiation, the molecular basis of apoptosis is less well understood. Genes involved in both cell proliferation and apoptosis reflect the role of some stimuli in both of these processes, the cell response depending on the overall cellular milieu. Recent research has given fascinating insights into the complex genetic and molecular mechanisms regulating apoptosis. A picture is emerging of the initiation in certain cells, after an apoptotic trigger, of sequential gene expression and specific signal transduction cascades that guide cells along the cell death pathway. Changes in gene expression precede the better known biochemical and morphological changes of apoptosis. It seems possible that, as a result of increased understanding of the cellular events preceding cell death, apoptosis may become more amenable to manipulation by appropriate drug- and gene-based therapies.
Collapse
Affiliation(s)
- K S Saini
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland, Australia
| | | |
Collapse
|
38
|
Brennand DM, Scully MF, Kakkar VV, Patel G. A cyclic peptide analogue of loop III of PDGF-BB causes apoptosis in human fibroblasts. FEBS Lett 1997; 419:166-70. [PMID: 9428627 DOI: 10.1016/s0014-5793(97)01446-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A cyclic peptide analogue of platelet-derived growth factor-BB (PDGF-BB), P1 [77IVRKK81-C-73RKIE76], has recently been shown to inhibit specifically [125I]PDGF-BB/receptor binding, and PDGF-BB-induced DNA synthesis in cells expressing PDGF receptors. Here we demonstrate that P1 induces apoptosis in exponentially growing human fibroblasts as confirmed by characteristic changes in cell and nuclear morphology, by TUNEL staining and by flow cytometry. Following incubation with P1 (100 microM), the percentage of cells exhibiting DNA fragmentation increased from 24% after 8 h to 76% after 28 h as exponentially growing cells progressed through the cell cycle. We conclude from these findings taken together that apoptosis accounts for the major proportion of P1-induced cell death. Omission of the Cys residue from P1 or replacement by Ser did not alter the potency of the peptide confirming that peptide dimerisation is not important for its activity. PDGF-BB, EGF, FGF, thrombin and foetal bovine serum were not able to rescue cells from the effects of P1. P1 is a useful tool for investigation of the balance of cellular proliferation/apoptosis in wound healing, atherosclerosis and restenosis, and constitutes a basis from which to design compounds with greater potency.
Collapse
Affiliation(s)
- D M Brennand
- Leopold Muller Laboratory, Thrombosis Research Institute, Chelsea, London, UK
| | | | | | | |
Collapse
|
39
|
Liu P, Ying Y, Anderson RG. Platelet-derived growth factor activates mitogen-activated protein kinase in isolated caveolae. Proc Natl Acad Sci U S A 1997; 94:13666-70. [PMID: 9391083 PMCID: PMC28363 DOI: 10.1073/pnas.94.25.13666] [Citation(s) in RCA: 186] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/1997] [Indexed: 02/05/2023] Open
Abstract
The ability of a peptide hormone to affect many different intracellular targets is thought to be possible because of the modular organization of signal transducing molecules in the cell. Evidence for the presence of signaling modules in metazoan cells, however, is incomplete. Herein we show, with morphology and cell fractionation, that all the components of a mitogen-activated protein kinase pathway are concentrated in caveolae of unstimulated human fibroblasts. Addition of platelet-derived growth factor to either the intact cell or caveolae isolated from these cells stimulates tyrosine phosphorylation and activates mitogen-activated protein kinases in caveolae. The molecular machinery for kinase activation, therefore, is preorganized at the cell surface of quiescent cells.
Collapse
Affiliation(s)
- P Liu
- Department of Cell Biology and Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75235-9039, USA
| | | | | |
Collapse
|
40
|
Funato N, Moriyama K, Shimokawa H, Kuroda T. Basic fibroblast growth factor induces apoptosis in myofibroblastic cells isolated from rat palatal mucosa. Biochem Biophys Res Commun 1997; 240:21-6. [PMID: 9367874 DOI: 10.1006/bbrc.1997.7588] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The effect of basic fibroblast growth factor (bFGF) on apoptosis in normal rat palatal fibroblasts and rat palatal scar fibroblasts was examined by the TUNEL method in order to clarify the mechanism of apoptosis induction in myofibroblasts during the scar formation process. A percentage of scar fibroblasts undergoing apoptosis was significantly higher than that of palatal fibroblasts when they were treated with bFGF succeeding to serum starvation. Palatal fibroblasts, phenotypically modulated into myofibroblasts by the pretreatment with transforming growth factor-beta 1 (TGF-beta 1), similarly showed a higher level of apoptosis induction by bFGF-treatment. TGF-beta 1 elevated protein and mRNA level of FGF receptor (FGFR) in palatal fibroblasts. Tyrosine autophosphorylation of FGFR upon stimulation by bFGF was significantly higher in scar fibroblasts than in normal palatal fibroblasts. These findings suggested that bFGF may be a potential stimulator of apoptosis in myofibroblasts during palatal scar formation and that FGFR may be responsible for this process.
Collapse
Affiliation(s)
- N Funato
- Department of Maxillo-Facial Orthognathics, Graduate School of Dentistry, Tokyo Medical and Dental University, Japan
| | | | | | | |
Collapse
|
41
|
Tateishi S, Yamaizumi M. Cell cycle control is aberrant in Chinese hamster ovary cell mutants exhibiting apoptosis after serum deprivation. SOMATIC CELL AND MOLECULAR GENETICS 1997; 23:313-23. [PMID: 9546075 DOI: 10.1007/bf02674279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We isolated mutants of Chinese hamster ovary cells that exhibit excessive apoptosis after serum deprivation. In the medium containing 10% serum, the growth rates of the mutants were 1.4 to 1.5-fold faster than those of wild-type cells. Whereas the cell cycle of wild-type cells was arrested at the G1 phase after serum deprivation, the cell cycle of the mutant cells was not fully arrested at this phase, suggesting that cell cycle regulation was disorganized in the mutants. The mutants were highly sensitive to a nucleotide-analogue 5-fluorouracil in the absence of serum, whereas wild-type cells were resistant to the drug. Based on the sensitivity to the drug after serum deprivation, we could classify the mutants into dominant groups and at least two recessive complementation groups. Thus, these mutants presumably contain different lesions in gene(s) required for cell cycle regulation and apoptosis.
Collapse
Affiliation(s)
- S Tateishi
- Institute of Molecular Embryology and Genetics, Kumamoto University School of Medicine, Japan
| | | |
Collapse
|
42
|
Abstract
BACKGROUND Prostate cancer is the most commonly diagnosed neoplasm and the second leading cause of male death in this country. Multiple genetic and epigenetic factors have been implicated in the oncogenesis and progression of prostate cancer. However, the molecular mechanisms underlying the disease remain largely unknown. The major difficulty in the clinical management of prostate cancer stems from the reality that reliable and accurate diagnostic/prognostic biomarkers are not available and that effective treatment regimens for hormone-resistant prostate cancers are yet to be developed. METHODS The present review, through extensive literature research, summarizes the most recently accumulated experimental and clinical data on the relationship between apoptosis and prostate cancer. We analyze the possibility of inducing prostate cancer cell apoptosis by: 1) androgen ablation by castration or biochemical antagonists: 2) chemotherapeutic drugs or natural/synthetic chemicals; 3) manipulation of apoptosis-related oncoproteins; and 4) modulation of intracellular signal transducers. RESULTS 1) Prostate cancer, like most other solid tumors, represents a very heterogeneous entity. Most prostate cancers, at the time of clinical diagnosis, present themselves as mixtures of androgen-dependent and androgen-independent cells. 2) Most prostate cancers respond initially to androgen ablation since the population of androgen-dependent cells undergoes rapid apoptosis upon androgen withdrawal. However, androgen ablation rarely cures patients, most of whom will experience recurrence due to takeover of the tumor mass by androgen-independent tumor cells as well as the emergence of apoptosis-resistant clones as a result of further genetic alterations such as bcl-2 amplification. 3) On the other hand, although androgen-independent prostate cancer cells do not undergo apoptosis upon androgen blocking, they do maintain the appropriate molecular machinery of apoptosis. Therefore, certain conventional chemotherapy drugs can eliminate androgen-independent cancer cells by inducing apoptosis. 4) However, most drugs used in chemotherapy induce apoptosis or mediate cytotoxicity only in proliferating cancer cells. Human prostate cancer cells demonstrate very slow growth kinetics. Thus, novel chemical/natural products need be identified to eradicate those nonproliferating cancer cells. In this regard, the angiogenesis inhibitor, linomide, and a plant extract, beta-lapachone, demonstrate very promising apoptosis-inducing effects on prostate cancer cells in a proliferation-independent manner. 5) An alternative way to modulate the apoptotic response is by interfering with the expression levels of essential regulatory molecule of apoptosis. Bcl-2 and p53 represent two prime targets for such manipulations. 6) Finally, modulation of signal transduction pathways (e.g., intracellular Ca2+ levels, PKC activity) involved in apoptosis may also induce and/or enhance the apoptotic response of prostate cancer cells. CONCLUSIONS Modulation of apoptotic response represents a novel mechanism-based approach which may help identify novel drugs and/or develop new therapeutic regimens for the treatment of prostate cancers.
Collapse
Affiliation(s)
- D G Tang
- Department of Radiation Oncology, Wayne State University, Detroit, Michigan 48202, USA.
| | | |
Collapse
|
43
|
Chin YE, Kitagawa M, Kuida K, Flavell RA, Fu XY. Activation of the STAT signaling pathway can cause expression of caspase 1 and apoptosis. Mol Cell Biol 1997; 17:5328-37. [PMID: 9271410 PMCID: PMC232383 DOI: 10.1128/mcb.17.9.5328] [Citation(s) in RCA: 404] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Protein tyrosine kinases activate the STAT (signal transducer and activator of transcription) signaling pathway, which can play essential roles in cell differentiation, cell cycle control, and development. However, the potential role of the STAT signaling pathway in the induction of apoptosis remains unexplored. Here we show that gamma interferon (IFN-gamma) activated STAT1 and induced apoptosis in both A431 and HeLa cells, whereas epidermal growth factor (EGF) activated STAT proteins and induced apoptosis in A431 but not in HeLa cells. EGF receptor autophosphorylation and mitogen-activated protein kinase activation in response to EGF were similar in both cell lines. The breast cancer cell line MDA-MB-468 exhibited a similar response to A431 cells, i.e., STAT activation and apoptosis correlatively resulted from EGF or IFN-gamma treatment. In addition, in a mutant A431 cell line in which STAT activation was abolished, no apoptosis was induced by either EGF or IFN-gamma. We further demonstrated that both EGF and IFN-gamma induced caspase 1 (interleukin-1beta converting enzyme [ICE]) gene expression in a STAT-dependent manner. IFN-gamma was unable to induce ICE gene expression and apoptosis in either JAK1-deficient HeLa cells (E2A4) or STAT1-deficient cells (U3A). However, ICE gene expression and apoptosis were induced by IFN-gamma in U3A cells into which STAT1 had been reintroduced. Moreover, both EGF-induced apoptosis and IFN-gamma-induced apoptosis were effectively blocked by Z-Val-Ala-Asp-fluoromethylketone (ZVAD) in all the cells tested, and studies from ICE-deficient cells indicated that ICE gene expression was necessary for IFN-gamma-induced apoptosis. We conclude that activation of the STAT signaling pathway can induce apoptosis through the induction of ICE gene expression.
Collapse
Affiliation(s)
- Y E Chin
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520-8023, USA
| | | | | | | | | |
Collapse
|
44
|
He J, deCastro CM, Vandenbark GR, Busciglio J, Gabuzda D. Astrocyte apoptosis induced by HIV-1 transactivation of the c-kit protooncogene. Proc Natl Acad Sci U S A 1997; 94:3954-9. [PMID: 9108086 PMCID: PMC20549 DOI: 10.1073/pnas.94.8.3954] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
HIV-1 infection of the central nervous system (CNS) frequently causes dementia and other neurological disorders. The mechanisms of CNS injury in HIV-1 infection are poorly understood. Apoptosis of neurons and astrocytes is induced by HIV-1 infection in vitro and in brain tissue from AIDS patients, but the apoptotic stimuli have not been identified. We report herein that HIV-1 infection of primary brain cultures induces the receptor tyrosine kinase protooncogene c-kit and that high levels of c-Kit expression are associated with astrocyte apoptosis. Overexpression of c-Kit in an astrocyte-derived cell line in the absence of HIV-1 induces rapid apoptotic death. The apoptotic mechanism requires the c-Kit tyrosine kinase domain. The mechanism of c-kit induction by HIV-1 involves transactivation of the c-kit promoter by the HIV-1 Nef protein. These studies demonstrate that c-Kit can induce astrocyte apoptosis and suggest that this mechanism may play a role in CNS injury caused by HIV-1 infection. We propose that c-Kit can serve dual functions as a growth factor receptor or apoptosis inducer.
Collapse
Affiliation(s)
- J He
- Dana-Farber Cancer Institute, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
45
|
Xu Y, Nguyen Q, Lo DC, Czaja MJ. c-myc-dependent hepatoma cell apoptosis results from oxidative stress and not a deficiency of growth factors. J Cell Physiol 1997; 170:192-9. [PMID: 9009148 DOI: 10.1002/(sici)1097-4652(199702)170:2<192::aid-jcp11>3.0.co;2-k] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Expression of c-myc regulates apoptotic cell death in the human hepatoma cell line HuH-7 during culture in serum-free medium (SFM) plus zinc. To understand the mechanism of this c-myc effect, the ability of various serum-contained factors to prevent apoptosis was determined. Apoptosis was not inhibited by growth factors and was even accelerated by supplementation with insulin-like growth factor I or insulin. Cell death was prevented by SFM supplementation with the amino acid glutamine but not serine or asparagine. Improved cell survival with glutamine was associated with increased levels of glutathione (GSH). In HuH-7 cells cultured in SFM plus zinc, c-myc expression led to decreased levels of GSH, and elevated intracellular levels of hydrogen peroxide (H2O2). Cell death induced by c-myc expression was inhibited by the addition of catalase or dimethyl sulfoxide, a hydroxyl radical scavenger, or by increased intracellular expression of catalase. In contrast to findings in fibroblasts, c-myc-dependent apoptosis during serum deprivation in HuH-7 hepatoma cells was unrelated to a loss of growth factors. Apoptosis resulted from H2O2-mediated oxidative stress with associated glutamine dependent intracellular GSH depletion.
Collapse
Affiliation(s)
- Y Xu
- Department of Medicine and the Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | |
Collapse
|
46
|
Buttyan R, Gobé G. Apoptosis in the mammalian kidney: incidence, effectors, and molecular control in normal development and disease states. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1997; 41:369-81. [PMID: 9204152 DOI: 10.1016/s1054-3589(08)61065-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In the preceding sections we have emphasized the current status of our knowledge concerning the involvement of apoptosis in normal and abnormal renal developmental processes, in control of the adult kidney size and capacity, in the development of renal disease states and in renal oncogenesis. At several points, we noted that studies of apoptosis in the kidney and in renal cells lag behind those in other organ systems. Even with the rudimentary knowledge now available, however, it is apparent that apoptosis is an extremely important process in the kidney. Recent observations lend credence to the view that continued study of this unique cell death process might enable the generation of novel and more effective therapies to treat renal diseases and renal malignancies. We wish to highlight several areas that require particular attention. First, the relationship between blood supply and apoptosis in the kidney requires further investigation. Benign human renal diseases are common in our population; and we now know that most of these diseases are associated with abnormal rates of apoptosis. Although the initiating agents for the various renal diseases vary, there is good reason to believe that much of the apoptosis that occurs in these adult diseases is the end result of reduced renal blood flow initiated by the causative agent. Cytokines or other extrinsic agents that can reduce the apoptotic loss of renal cells under these conditions hold theoretical promise in treating these diseases. Second, there is an urgent need to define the endocrine, paracrine, or autocrine roles of cytokines in normal renal physiology and in the pathogenesis of various renal syndromes. As indicated above, elaboration of fibrous extracellular material by fibroblasts in the tubulointerstitial regions of the kidney appears to be part of the final common pathway leading to end-stage renal disease. It is important to understand how the function of these fibroblasts is controlled. Conversely, apoptosis of glomerular or renal tubular cells also appears to play a role in the development of many of these syndromes. There is already experimental and clinical evidence showing that IGF-1 and hepatocyte growth factor therapies can be useful in renal diseases [57, 58]. It remains to be determined how much of the usefulness of these cytokines is related to their ability to suppress apoptosis as opposed to their ability to promote true growth. Finally, the analysis of apoptotic regulation during renal oncogenesis is critical. Maligant renal cell cancers are difficult to detect in adults before their metastases cause symptoms; and by this late stage renal tumors are almost invariably fatal. The ability of these tumors to regress spontaneously indicates that most apoptotic pathways are retained in these cells, yet their disappointing response to chemotherapy indicates that we have much to learn about how to trigger these pathways. Hopefully a better understanding of the control of these pathways will lead to improved therapy for this devastating group of neoplasms.
Collapse
Affiliation(s)
- R Buttyan
- Department of Urology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | | |
Collapse
|
47
|
Polunovsky VA, Rosenwald IB, Tan AT, White J, Chiang L, Sonenberg N, Bitterman PB. Translational control of programmed cell death: eukaryotic translation initiation factor 4E blocks apoptosis in growth-factor-restricted fibroblasts with physiologically expressed or deregulated Myc. Mol Cell Biol 1996; 16:6573-81. [PMID: 8887686 PMCID: PMC231659 DOI: 10.1128/mcb.16.11.6573] [Citation(s) in RCA: 135] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
There is increasing evidence that cell cycle transit is potentially lethal, with survival depending on the activation of metabolic pathways which block apoptosis. However, the identities of those pathways coupling cell cycle transit to survival remain undefined. Here we show that the eukaryotic translation initiation factor 4E (eIF4E) can mediate both proliferative and survival signaling. Overexpression of eIF4E completely substituted for serum or individual growth factors in preserving the viability of established NIH 3T3 fibroblasts. An eIF4E mutant (Ser-53 changed to Ala) defective in mediating its growth-factor-regulated functions was also defective in its survival signaling. Survival signaling by enforced expression of eIF4E did not result from autocrine release of survival factors, nor did it lead to increased expression of the apoptosis antagonists Bcl-2 and Bcl-XL. In addition, the execution apparatus of the apoptotic response in eIF4E-overexpressing cells was found to be intact. Increased expression of eIF4E was sufficient to inhibit apoptosis in serum-restricted primary fibroblasts with enforced expression of Myc. In contrast, activation of Ha-Ras, which is required for eIF4E proliferative signaling, did not suppress Myc-induced apoptosis. These data suggest that the eIF4E-activated pathways leading to survival and cell cycle progression are distinct. This dual signaling of proliferation and survival might be the basis for the potency of eIF4E as an inducer of neoplastic transformation.
Collapse
Affiliation(s)
- V A Polunovsky
- Pulmonary and Critical Care Division, Department of Medicine, University of Minnesota Medical School, Minneapolis 55455, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Wang ZY, Deuel TF. S1-nuclease-sensitive DNA structures contribute to transcriptional regulation of the human PDGF A-chain. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 1996; 55:227-44. [PMID: 8787612 DOI: 10.1016/s0079-6603(08)60195-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Z Y Wang
- Department of Medicine and Biochemistry, Jewish Hospital at Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|