1
|
Camps-Fajol C, Cavero D, Minguillón J, Surrallés J. Targeting protein-protein interactions in drug discovery: Modulators approved or in clinical trials for cancer treatment. Pharmacol Res 2025; 211:107544. [PMID: 39667542 DOI: 10.1016/j.phrs.2024.107544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
Protein-protein interactions (PPIs) form complex cellular networks fundamental to many key biological processes, including signal transduction, cell proliferation and DNA repair. In consequence, their perturbation is often associated with many human diseases. Targeting PPIs offers a promising approach in drug discovery and ongoing advancements in this field hold the potential to provide highly specific therapies for a wide range of complex diseases. Despite the development of PPI modulators is challenging, advances in the genetic, proteomic and computational level have facilitated their discovery and optimization. Focusing on anticancer drugs, in the last years several PPI modulators have entered clinical trials and venetoclax, which targets Bcl-2 family proteins, has been approved for treating different types of leukemia. This review discusses the clinical development status of drugs modulating several PPIs, such as MDM2-4/p53, Hsp90/Hsp90, Hsp90/CDC37, c-Myc/Max, KRAS/SOS1, CCR5/CCL5, CCR2/CCL2 or Smac/XIAP, in cancer drug discovery.
Collapse
Affiliation(s)
- Cristina Camps-Fajol
- Unitat Mixta de Recerca en Medicina Genòmica, Universitat Autònoma de Barcelona (UAB)-IR SANT PAU, Barcelona, Spain; Institut de Bioenginyeria de Catalunya (IBEC), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CIBERER, ISCIII), Madrid, Spain
| | - Debora Cavero
- Unitat Mixta de Recerca en Medicina Genòmica, Universitat Autònoma de Barcelona (UAB)-IR SANT PAU, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CIBERER, ISCIII), Madrid, Spain
| | - Jordi Minguillón
- CIBERER-ISCIII, IdiPAZ-CNIO Translational Research Unit in Pediatric Hemato-Oncology, La Paz University Hospital Research Institute; Spanish National Cancer Center, Madrid, Spain; Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Jordi Surrallés
- Unitat Mixta de Recerca en Medicina Genòmica, Universitat Autònoma de Barcelona (UAB)-IR SANT PAU, Barcelona, Spain; Institut de Bioenginyeria de Catalunya (IBEC), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CIBERER, ISCIII), Madrid, Spain; Servei de Genètica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Spain.
| |
Collapse
|
2
|
Lafita-Navarro MC, Liaño-Pons J, Quintanilla A, Varela I, Blanco R, Ourique F, Bretones G, Aresti J, Molina E, Carroll P, Hurlin P, Romero OA, Sanchez-Céspedes M, Eisenman RN, Delgado MD, León J. The MNT transcription factor autoregulates its expression and supports proliferation in MYC-associated factor X (MAX)-deficient cells. J Biol Chem 2020; 295:2001-2017. [PMID: 31919096 PMCID: PMC7029127 DOI: 10.1074/jbc.ra119.010389] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
The MAX network transcriptional repressor (MNT) is an MXD family transcription factor of the basic helix-loop-helix (bHLH) family. MNT dimerizes with another transcriptional regulator, MYC-associated factor X (MAX), and down-regulates genes by binding to E-boxes. MAX also dimerizes with MYC, an oncogenic bHLH transcription factor. Upon E-box binding, the MYC-MAX dimer activates gene expression. MNT also binds to the MAX dimerization protein MLX (MLX), and MNT-MLX and MNT-MAX dimers co-exist. However, all MNT functions have been attributed to MNT-MAX dimers, and no functions of the MNT-MLX dimer have been described. MNT's biological role has been linked to its function as a MYC oncogene modulator, but little is known about its regulation. We show here that MNT localizes to the nucleus of MAX-expressing cells and that MNT-MAX dimers bind and repress the MNT promoter, an effect that depends on one of the two E-boxes on this promoter. In MAX-deficient cells, MNT was overexpressed and redistributed to the cytoplasm. Interestingly, MNT was required for cell proliferation even in the absence of MAX. We show that in MAX-deficient cells, MNT binds to MLX, but also forms homodimers. RNA-sequencing experiments revealed that MNT regulates the expression of several genes even in the absence of MAX, with many of these genes being involved in cell cycle regulation and DNA repair. Of note, MNT-MNT homodimers regulated the transcription of some genes involved in cell proliferation. The tight regulation of MNT and its functionality even without MAX suggest a major role for MNT in cell proliferation.
Collapse
Affiliation(s)
- M Carmen Lafita-Navarro
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Judit Liaño-Pons
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Andrea Quintanilla
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Ignacio Varela
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Rosa Blanco
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Fabiana Ourique
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Gabriel Bretones
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Julia Aresti
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Ester Molina
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Patrick Carroll
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Peter Hurlin
- Shriners Hospitals for Children Research Center, and Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239
| | - Octavio A Romero
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute-IDIBELL, 08908 Barcelona, Spain
| | - Montse Sanchez-Céspedes
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute-IDIBELL, 08908 Barcelona, Spain
| | - Robert N Eisenman
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - M Dolores Delgado
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC, and Department of Molecular Biology, Universidad de Cantabria, 39005 Santander, Spain.
| |
Collapse
|
3
|
Yesudhas D, Batool M, Anwar MA, Panneerselvam S, Choi S. Proteins Recognizing DNA: Structural Uniqueness and Versatility of DNA-Binding Domains in Stem Cell Transcription Factors. Genes (Basel) 2017; 8:genes8080192. [PMID: 28763006 PMCID: PMC5575656 DOI: 10.3390/genes8080192] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/22/2017] [Accepted: 07/25/2017] [Indexed: 12/17/2022] Open
Abstract
Proteins in the form of transcription factors (TFs) bind to specific DNA sites that regulate cell growth, differentiation, and cell development. The interactions between proteins and DNA are important toward maintaining and expressing genetic information. Without knowing TFs structures and DNA-binding properties, it is difficult to completely understand the mechanisms by which genetic information is transferred between DNA and proteins. The increasing availability of structural data on protein-DNA complexes and recognition mechanisms provides deeper insights into the nature of protein-DNA interactions and therefore, allows their manipulation. TFs utilize different mechanisms to recognize their cognate DNA (direct and indirect readouts). In this review, we focus on these recognition mechanisms as well as on the analysis of the DNA-binding domains of stem cell TFs, discussing the relative role of various amino acids toward facilitating such interactions. Unveiling such mechanisms will improve our understanding of the molecular pathways through which TFs are involved in repressing and activating gene expression.
Collapse
Affiliation(s)
- Dhanusha Yesudhas
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea.
| | - Maria Batool
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea.
| | - Muhammad Ayaz Anwar
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea.
| | - Suresh Panneerselvam
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea.
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea.
| |
Collapse
|
4
|
Allevato M, Bolotin E, Grossman M, Mane-Padros D, Sladek FM, Martinez E. Sequence-specific DNA binding by MYC/MAX to low-affinity non-E-box motifs. PLoS One 2017; 12:e0180147. [PMID: 28719624 PMCID: PMC5515408 DOI: 10.1371/journal.pone.0180147] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/09/2017] [Indexed: 01/07/2023] Open
Abstract
The MYC oncoprotein regulates transcription of a large fraction of the genome as an obligatory heterodimer with the transcription factor MAX. The MYC:MAX heterodimer and MAX:MAX homodimer (hereafter MYC/MAX) bind Enhancer box (E-box) DNA elements (CANNTG) and have the greatest affinity for the canonical MYC E-box (CME) CACGTG. However, MYC:MAX also recognizes E-box variants and was reported to bind DNA in a “non-specific” fashion in vitro and in vivo. Here, in order to identify potential additional non-canonical binding sites for MYC/MAX, we employed high throughput in vitro protein-binding microarrays, along with electrophoretic mobility-shift assays and bioinformatic analyses of MYC-bound genomic loci in vivo. We identified all hexameric motifs preferentially bound by MYC/MAX in vitro, which include the low-affinity non-E-box sequence AACGTT, and found that the vast majority (87%) of MYC-bound genomic sites in a human B cell line contain at least one of the top 21 motifs bound by MYC:MAX in vitro. We further show that high MYC/MAX concentrations are needed for specific binding to the low-affinity sequence AACGTT in vitro and that elevated MYC levels in vivo more markedly increase the occupancy of AACGTT sites relative to CME sites, especially at distal intergenic and intragenic loci. Hence, MYC binds diverse DNA motifs with a broad range of affinities in a sequence-specific and dose-dependent manner, suggesting that MYC overexpression has more selective effects on the tumor transcriptome than previously thought.
Collapse
Affiliation(s)
- Michael Allevato
- Department of Biochemistry, University of California Riverside, Riverside, California, United States of America
| | - Eugene Bolotin
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California, United States of America
| | - Mark Grossman
- Department of Biochemistry, University of California Riverside, Riverside, California, United States of America
| | - Daniel Mane-Padros
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California, United States of America
| | - Frances M. Sladek
- Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California, United States of America
- * E-mail: (E.M.); (F.M.S.)
| | - Ernest Martinez
- Department of Biochemistry, University of California Riverside, Riverside, California, United States of America
- * E-mail: (E.M.); (F.M.S.)
| |
Collapse
|
5
|
Talamillo A, Grande L, Ruiz-Ontañon P, Velasquez C, Mollinedo P, Torices S, Sanchez-Gomez P, Aznar A, Esparis-Ogando A, Lopez-Lopez C, Lafita C, Berciano MT, Montero JA, Vazquez-Barquero A, Segura V, Villagra NT, Pandiella A, Lafarga M, Leon J, Martinez-Climent JA, Sanz-Moreno V, Fernandez-Luna JL. ODZ1 allows glioblastoma to sustain invasiveness through a Myc-dependent transcriptional upregulation of RhoA. Oncogene 2017; 36:1733-1744. [PMID: 27641332 DOI: 10.1038/onc.2016.341] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 08/01/2016] [Accepted: 08/10/2016] [Indexed: 02/06/2023]
Abstract
Long-term survival remains low for most patients with glioblastoma (GBM), which reveals the need for markers of disease outcome and novel therapeutic targets. We describe that ODZ1 (also known as TENM1), a type II transmembrane protein involved in fetal brain development, plays a crucial role in the invasion of GBM cells. Differentiation of glioblastoma stem-like cells drives the nuclear translocation of an intracellular fragment of ODZ1 through proteolytic cleavage by signal peptide peptidase-like 2a. The intracellular fragment of ODZ1 promotes cytoskeletal remodelling of GBM cells and invasion of the surrounding environment both in vitro and in vivo. Absence of ODZ1 by gene deletion or downregulation of ODZ1 by small interfering RNAs drastically reduces the invasive capacity of GBM cells. This activity is mediated by an ODZ1-triggered transcriptional pathway, through the E-box binding Myc protein, that promotes the expression and activation of Ras homolog family member A (RhoA) and subsequent activation of Rho-associated, coiled-coil containing protein kinase (ROCK). Overexpression of ODZ1 in GBM cells reduced survival of xenografted mice. Consistently, analysis of 122 GBM tumour samples revealed that the number of ODZ1-positive cells inversely correlated with overall and progression-free survival. Our findings establish a novel marker of invading GBM cells and consequently a potential marker of disease progression and a therapeutic target in GBM.
Collapse
Affiliation(s)
- A Talamillo
- Unidad de Genética, Hospital Valdecilla-IDIVAL, Santander, Spain
| | - L Grande
- Unidad de Genética, Hospital Valdecilla-IDIVAL, Santander, Spain
| | - P Ruiz-Ontañon
- Unidad de Genética, Hospital Valdecilla-IDIVAL, Santander, Spain
| | - C Velasquez
- Servicio de Neurocirugía, Hospital Valdecilla-IDIVAL, Santander, Spain
| | - P Mollinedo
- Unidad de Genética, Hospital Valdecilla-IDIVAL, Santander, Spain
| | - S Torices
- Unidad de Genética, Hospital Valdecilla-IDIVAL, Santander, Spain
| | - P Sanchez-Gomez
- Unidad de Neuro-Oncología, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - A Aznar
- Centro para la Investigación Médica Aplicada (CIMA), Pamplona, Spain
| | - A Esparis-Ogando
- Centro de Investigación del Cáncer (CSIC-USAL), Salamanca, Spain
| | - C Lopez-Lopez
- Servicio de Oncología Médica, Hospital Valdecilla-IDIVAL, Santander, Spain
| | - C Lafita
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria, Santander, Spain
| | - M T Berciano
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria, Santander, Spain
| | - J A Montero
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria, Santander, Spain
| | | | - V Segura
- Centro para la Investigación Médica Aplicada (CIMA), Pamplona, Spain
| | - N T Villagra
- Servicio de Anatomía Patológica, Hospital Valdecilla and Instituto de Investigación Valdecilla (IDIVAL), Santander, Spain
| | - A Pandiella
- Centro de Investigación del Cáncer (CSIC-USAL), Salamanca, Spain
| | - M Lafarga
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria, Santander, Spain
| | - J Leon
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria, Santander, Spain
| | | | - V Sanz-Moreno
- Randall Division of Cell and Molecular Biophysics, School of Biomedical and Health Sciences, King's College London, London, UK
| | | |
Collapse
|
6
|
García-Alegría E, Lafita-Navarro MC, Aguado R, García-Gutiérrez L, Sarnataro K, Ruiz-Herguido C, Martín F, Bigas A, Canelles M, León J. NUMB inactivation confers resistance to imatinib in chronic myeloid leukemia cells. Cancer Lett 2016; 375:92-99. [PMID: 26944313 DOI: 10.1016/j.canlet.2016.02.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 01/21/2023]
Abstract
Chronic myeloid leukemia (CML) progresses from a chronic to a blastic phase, where the leukemic cells are proliferative and undifferentiated. The CML is nowadays successfully treated with BCR-ABL kinase inhibitors as imatinib and its derivatives. NUMB is an evolutionary well-conserved protein initially described as a functional antagonist of NOTCH function. NUMB is an endocytic protein associated with receptor internalization, involved in multiple cellular functions. It has been reported that MSI2 protein, a NUMB inhibitor, is upregulated in CML blast crisis, whereas NUMB itself is downregulated. This suggest that NUMB plays a role in the malignant progression of CML. Here we have generated K562 cells (derived from CML in blast crisis) constitutively expressing a dominant negative form of NUMB (dnNUMB). We show that dnNUMB expression confers a high proliferative phenotype to the cells. Importantly, dnNUMB triggers a partial resistance to imatinib in these cells, antagonizing the apoptosis mediated by the drug. Interestingly, imatinib resistance is not linked to p53 status or NOTCH signaling, as K562 lack p53 and imatinib resistance is reproduced in the presence of NOTCH inhibitors. Taken together, our data support the hypothesis that NUMB activation could be a new therapeutic target in CML.
Collapse
Affiliation(s)
- Eva García-Alegría
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - M Carmen Lafita-Navarro
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Rocío Aguado
- Instituto de Parasitología y Biomedicina, CSIC, P. T. Ciencias de la Salud, Granada, Spain
| | - Lucia García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Kyle Sarnataro
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | | | | | - Anna Bigas
- Stem Cells and Cancer Group. IMIM, Barcelona, Spain
| | - Matilde Canelles
- Instituto de Parasitología y Biomedicina, CSIC, P. T. Ciencias de la Salud, Granada, Spain.
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria and Dpto. de Biología Molecular, Universidad de Cantabria, Santander, Spain.
| |
Collapse
|
7
|
Sharma T, Bansal R, Haokip DT, Goel I, Muthuswami R. SMARCAL1 Negatively Regulates C-Myc Transcription By Altering The Conformation Of The Promoter Region. Sci Rep 2015; 5:17910. [PMID: 26648259 PMCID: PMC4673416 DOI: 10.1038/srep17910] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022] Open
Abstract
SMARCAL1, a member of the SWI2/SNF2 protein family, stabilizes replication forks during DNA damage. In this manuscript, we provide the first evidence that SMARCAL1 is also a transcriptional co-regulator modulating the expression of c-Myc, a transcription factor that regulates 10-15% genes in the human genome. BRG1, SMARCAL1 and RNAPII were found localized onto the c-myc promoter. When HeLa cells were serum starved, the occupancy of SMARCAL1 on the c-myc promoter increased while that of BRG1 and RNAPII decreased correlating with repression of c-myc transcription. Using Active DNA-dependent ATPase A Domain (ADAAD), the bovine homolog of SMARCAL1, we show that the protein can hydrolyze ATP using a specific region upstream of the CT element of the c-myc promoter as a DNA effector. The energy, thereby, released is harnessed to alter the conformation of the promoter DNA. We propose that SMARCAL1 negatively regulates c-myc transcription by altering the conformation of its promoter region during differentiation.
Collapse
Affiliation(s)
| | - Ritu Bansal
- School of Life Sciences, JNU, New Delhi 110067
| | | | - Isha Goel
- School of Life Sciences, JNU, New Delhi 110067
| | | |
Collapse
|
8
|
Growth-promoting and tumourigenic activity of c-Myc is suppressed by Hhex. Oncogene 2014; 34:3011-22. [PMID: 25220416 DOI: 10.1038/onc.2014.240] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 06/18/2014] [Accepted: 06/30/2014] [Indexed: 12/12/2022]
Abstract
c-Myc transcription factor is a key protein involved in cellular growth, proliferation and metabolism. c-Myc is one of the most frequently activated oncogenes, highlighting the need to identify intracellular molecules that interact directly with c-Myc to suppress its function. Here we show that Hhex is able to interact with the basic region/helix-loop-helix/leucine zipper of c-Myc. Knockdown of Hhex increases proliferation rate in hepatocellular carcinoma cells, whereas Hhex expression cell-autonomously reduces cell proliferation rate in multiple cell lines by increasing G1 phase length through a c-Myc-dependent mechanism. Global transcriptomic analysis shows that Hhex counter-regulates multiple c-Myc targets involved in cell proliferation and metabolism. Concomitantly, Hhex expression leads to reduced cell size, lower levels of cellular RNA, downregulation of metabolism-related genes, decreased sensitivity to methotrexate and severe reduction in the ability to form tumours in nude mouse xenografts, all indicative of decreased c-Myc activity. Our data suggest that Hhex is a novel regulator of c-Myc function that limits c-Myc activity in transformed cells.
Collapse
|
9
|
Conacci-Sorrell M, McFerrin L, Eisenman RN. An overview of MYC and its interactome. Cold Spring Harb Perspect Med 2014; 4:a014357. [PMID: 24384812 DOI: 10.1101/cshperspect.a014357] [Citation(s) in RCA: 326] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This review is intended to provide a broad outline of the biological and molecular functions of MYC as well as of the larger protein network within which MYC operates. We present a view of MYC as a sensor that integrates multiple cellular signals to mediate a broad transcriptional response controlling many aspects of cell behavior. We also describe the larger transcriptional network linked to MYC with emphasis on the MXD family of MYC antagonists. Last, we discuss evidence that the network has evolved for millions of years, dating back to the emergence of animals.
Collapse
|
10
|
Reverse engineering the neuroblastoma regulatory network uncovers MAX as one of the master regulators of tumor progression. PLoS One 2013; 8:e82457. [PMID: 24349289 PMCID: PMC3857773 DOI: 10.1371/journal.pone.0082457] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/23/2013] [Indexed: 12/17/2022] Open
Abstract
Neuroblastoma is the most common extracranial tumor and a major cause of infant cancer mortality worldwide. Despite its importance, little is known about its molecular mechanisms. A striking feature of this tumor is its clinical heterogeneity. Possible outcomes range from aggressive invasion to other tissues, causing patient death, to spontaneous disease regression or differentiation into benign ganglioneuromas. Several efforts have been made in order to find tumor progression markers. In this work, we have reconstructed the neuroblastoma regulatory network using an information-theoretic approach in order to find genes involved in tumor progression and that could be used as outcome predictors or as therapeutic targets. We have queried the reconstructed neuroblastoma regulatory network using an aggressive neuroblastoma metastasis gene signature in order to find its master regulators (MRs). MRs expression profiles were then investigated in other neuroblastoma datasets so as to detect possible clinical significance. Our analysis pointed MAX as one of the MRs of neuroblastoma progression. We have found that higher MAX expression correlated with favorable patient outcomes. We have also found that MAX expression and protein levels were increased during neuroblastoma SH-SY5Y cells differentiation. We propose that MAX is involved in neuroblastoma progression, possibly increasing cell differentiation by means of regulating the availability of MYC:MAX heterodimers. This mechanism is consistent with the results found in our SH-SY5Y differentiation protocol, suggesting that MAX has a more central role in these cells differentiation than previously reported. Overexpression of MAX has been identified as anti-tumorigenic in other works, but, to our knowledge, this is the first time that the link between the expression of this gene and malignancy was verified under physiological conditions.
Collapse
|
11
|
Funakoshi-Tago M, Sumi K, Kasahara T, Tago K. Critical roles of Myc-ODC axis in the cellular transformation induced by myeloproliferative neoplasm-associated JAK2 V617F mutant. PLoS One 2013; 8:e52844. [PMID: 23300995 PMCID: PMC3536786 DOI: 10.1371/journal.pone.0052844] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 11/21/2012] [Indexed: 12/13/2022] Open
Abstract
The acquired mutation (V617F) of Janus kinase 2 (JAK2) is observed in the majority of patients with myeloproliferative neoplasms (MPNs). In the screening of genes whose expression was induced by JAK2 (V617F), we found the significant induction of c-Myc mRNA expression mediated by STAT5 activation. Interestingly, GSK-3β was inactivated in transformed Ba/F3 cells by JAK2 (V617F), and this enhanced the protein expression of c-Myc. The enforced expression of c-Myc accelerated cell proliferation but failed to inhibit apoptotic cell death caused by growth factor deprivation; however, the inhibition of GSK-3β completely inhibited the apoptosis of cells expressing c-Myc. Strikingly, c-Myc T58A mutant exhibited higher proliferative activity in a growth-factor-independent manner; however, this mutant failed to induce apoptosis. In addition, knockdown of c-Myc significantly inhibited the proliferation of transformed cells by JAK2 (V617F), suggesting that c-Myc plays an important role in oncogenic activity of JAK2 (V617F). Furthermore, JAK2 (V617F) induced the expression of a target gene of c-Myc, ornithine decarboxylase (ODC), known as the rate-limiting enzyme in polyamine biosynthesis. An ODC inhibitor, difluoromethylornithine (DFMO), prevented the proliferation of transformed cells by JAK2 (V617F). Importantly, administration of DFMO effectively delayed tumor formation in nude mice inoculated with transformed cells by JAK2 (V617F), resulting in prolonged survival; therefore, ODC expression through c-Myc is a critical step for JAK2 (V617F)-induced transformation and DFMO could be used as effective therapy for MPNs.
Collapse
Affiliation(s)
- Megumi Funakoshi-Tago
- Department of Biochemistry, Faculty of Pharmacology, Keio University, Tokyo, Japan
- * E-mail: (MF-T); (KT)
| | - Kazuya Sumi
- Department of Biochemistry, Faculty of Pharmacology, Keio University, Tokyo, Japan
| | - Tadashi Kasahara
- Department of Biochemistry, Faculty of Pharmacology, Keio University, Tokyo, Japan
| | - Kenji Tago
- Division of Structural Biochemistry, Department of Biochemistry, Jichi Medical University, Shimotsuke-shi, Japan
- * E-mail: (MF-T); (KT)
| |
Collapse
|
12
|
Abstract
Hematopoiesis is a process capable of generating millions of cells every second, as distributed in many cell types. The process is regulated by a number of transcription factors that regulate the differentiation along the distinct lineages and dictate the genetic program that defines each mature phenotype. Myc was first discovered as the oncogene of avian leukemogenic retroviruses; it was later found translocated in human lymphoma. From then on, evidence accumulated showing that c-Myc is one of the transcription factors playing a major role in hematopoiesis. The study of genetically modified mice with overexpression or deletion of Myc has shown that c-Myc is required for the correct balance between self-renewal and differentiation of hematopoietic stem cells (HSCs). Enforced Myc expression in mice leads to reduced HSC pools owing to loss of self-renewal activity at the expense of increased proliferation of progenitor cells and differentiation. c-Myc deficiency consistently results in the accumulation of HSCs. Other models with conditional Myc deletion have demonstrated that different lineages of hematopoietic cells differ in their requirement for c-Myc to regulate their proliferation and differentiation. When transgenic mice overexpress c-Myc or N-Myc in mature cells from the lymphoid or myeloid lineages, the result is lymphoma or leukemia. In agreement, enforced expression of c-Myc blocks the differentiation in several leukemia-derived cell lines capable of differentiating in culture. Not surprising, MYC deregulation is recurrently found in many types of human lymphoma and leukemia. Whereas MYC is deregulated by translocation in Burkitt lymphoma and, less frequently, other types of lymphoma, MYC is frequently overexpressed in acute lymphoblastic and myeloid leukemia, through mechanisms unrelated to chromosomal translocation, and is often associated with disease progression.
Collapse
Affiliation(s)
- M Dolores Delgado
- Departamento de Biología Molecular, Facultad de Medicina and Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | | |
Collapse
|
13
|
Albajar M, Gómez-Casares MT, Llorca J, Mauleon I, Vaqué JP, Acosta JC, Bermúdez A, Donato N, Delgado MD, León J. MYC in chronic myeloid leukemia: induction of aberrant DNA synthesis and association with poor response to imatinib. Mol Cancer Res 2011; 9:564-76. [PMID: 21460180 DOI: 10.1158/1541-7786.mcr-10-0356] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Untreated chronic myeloid leukemia (CML) progresses from chronic phase to blastic crisis (BC). Increased genomic instability, deregulated proliferation, and loss of differentiation appear associated to BC, but the molecular alterations underlying the progression of CML are poorly characterized. MYC oncogene is frequently deregulated in human cancer, often associated with tumor progression. Genomic instability and induction of aberrant DNA replication are described as effects of MYC. In this report, we studied MYC activities in CML cell lines with conditional MYC expression with and without exposure to imatinib, the front-line drug in CML therapy. In cells with conditional MYC expression, MYC did not rescue the proliferation arrest mediated by imatinib but provoked aberrant DNA synthesis and accumulation of cells with 4C content. We studied MYC mRNA expression in 66 CML patients at different phases of the disease, and we found that MYC expression was higher in CML patients at diagnosis than control bone marrows or in patients responding to imatinib. Further, high MYC levels at diagnosis correlated with a poor response to imatinib. MYC expression did not directly correlate with BCR-ABL levels in patients treated with imatinib. Overall our study suggests that, as in other tumor models, MYC-induced aberrant DNA synthesis in CML cells is consistent with MYC overexpression in untreated CML patients and nonresponding patients and supports a role for MYC in CML progression, possibly through promotion of genomic instability.
Collapse
Affiliation(s)
- Marta Albajar
- Departamento de Biología Molecular, Facultad de Medicina, Instituto de Biomedicina y Biotecnología de Cantabria, Avda Cardenal Herrera Oria s/n, 39011 Santander, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kassouf MT, Hughes JR, Taylor S, McGowan SJ, Soneji S, Green AL, Vyas P, Porcher C. Genome-wide identification of TAL1's functional targets: insights into its mechanisms of action in primary erythroid cells. Genome Res 2010; 20:1064-83. [PMID: 20566737 PMCID: PMC2909570 DOI: 10.1101/gr.104935.110] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 05/19/2010] [Indexed: 12/19/2022]
Abstract
Coordination of cellular processes through the establishment of tissue-specific gene expression programs is essential for lineage maturation. The basic helix-loop-helix hemopoietic transcriptional regulator TAL1 (formerly SCL) is required for terminal differentiation of red blood cells. To gain insight into TAL1 function and mechanisms of action in erythropoiesis, we performed ChIP-sequencing and gene expression analyses from primary fetal liver erythroid cells. We show that TAL1 coordinates expression of genes in most known red cell-specific processes. The majority of TAL1's genomic targets require direct DNA-binding activity. However, one-fifth of TAL1's target sequences, mainly among those showing high affinity for TAL1, can recruit the factor independently of its DNA binding activity. An unbiased DNA motif search of sequences bound by TAL1 identified CAGNTG as TAL1-preferred E-box motif in erythroid cells. Novel motifs were also characterized that may help distinguish activated from repressed genes and suggest a new mechanism by which TAL1 may be recruited to DNA. Finally, analysis of recruitment of GATA1, a protein partner of TAL1, to sequences occupied by TAL1 suggests that TAL1's binding is necessary prior or simultaneous to that of GATA1. This work provides the framework to study regulatory networks leading to erythroid terminal maturation and to model mechanisms of action of tissue-specific transcription factors.
Collapse
Affiliation(s)
- Mira T. Kassouf
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Jim R. Hughes
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Stephen Taylor
- Computational Biology Research Group (CBRG), Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Simon J. McGowan
- Computational Biology Research Group (CBRG), Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Shamit Soneji
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Angela L. Green
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Paresh Vyas
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Catherine Porcher
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, United Kingdom
| |
Collapse
|
15
|
Qu B, Wang W, Tan Z, Li D, Wan J, Sun J, Cheng K, Luo H. Diethyl (6-amino-9H-purin-9-yl) methylphosphonate induces apoptosis and cell cycle arrest in hepatocellular carcinoma BEL-7402 cells: Role of reactive oxygen species. Free Radic Res 2010; 44:881-90. [PMID: 20528564 DOI: 10.3109/10715762.2010.487868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The primary purpose of this work was to study the mechanism of the anti-proliferation activity of compound diethyl (6-amino-9H-purin-9-yl) methylphosphonate (DaMP), a novel acyclic nucleoside phosphonate. Using cell survival MTT assay, flow cytometry analysis, DNA laddering, DCF fluorescence detection and caspases assays, this study investigated the effects of this compound on cell apoptosis, cell cycle regulation and reactive oxygen species in human hepatocarcinoma BEL-7402 cell lines. Exposure to DaMP at 80 microM for 24 h, BEL-7402 cells displayed a marked retardation of S-phase progression, leading to a severe perturbation of normal cell cycle. In addition, DaMP also significantly inhibited cell proliferation by inducing apoptosis, disrupting DNA synthesis and increasing the activities of caspase-3 and -9, while the antioxidants could significantly inhibit these effects. This study was the first to demonstrate that DaMP could induce apoptosis and cell cycle arrest by producing reactive oxygen species and activating caspase-3 and -9.
Collapse
Affiliation(s)
- Bin Qu
- College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, Shandong, 266042, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Albihn A, Johnsen JI, Henriksson MA. MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res 2010; 107:163-224. [PMID: 20399964 DOI: 10.1016/s0065-230x(10)07006-5] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
MYC proteins (c-MYC, MYCN, and MYCL) regulate processes involved in many if not all aspects of cell fate. Therefore, it is not surprising that the MYC genes are deregulated in several human neoplasias as a result from genetic and epigenetic alterations. The near "omnipotency" together with the many levels of regulation makes MYC an attractive target for tumor intervention therapy. Here, we summarize some of the current understanding of MYC function and provide an overview of different cancer forms with MYC deregulation. We also describe available treatments and highlight novel approaches in the pursuit for MYC-targeting therapies. These efforts, at different stages of development, constitute a promising platform for novel, more specific treatments with fewer side effects. If successful a MYC-targeting therapy has the potential for tailored treatment of a large number of different tumors.
Collapse
Affiliation(s)
- Ami Albihn
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
17
|
Zhang B, Ma JX. SERPINA3K prevents oxidative stress induced necrotic cell death by inhibiting calcium overload. PLoS One 2008; 3:e4077. [PMID: 19115003 PMCID: PMC2605247 DOI: 10.1371/journal.pone.0004077] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Accepted: 11/27/2008] [Indexed: 11/28/2022] Open
Abstract
Background SERPINA3K, an extracellular serine proteinase inhibitor (serpin), has been shown to have decreased levels in the retinas of diabetic rats, which may contribute to diabetic retinopathy. The function of SERPINA3K in the retina has not been investigated. Methodology/Principal Findings The present study identified a novel function of SERPINA3K, i.e. it protects retinal cells against oxidative stress-induced cell death including retinal neuronal cells and Müller cells. Flow-cytometry showed that the protective effect of SERPINA3K on Müller cells is via reducing oxidation-induced necrosis. Measurements of intracellular calcium concentration showed that SERPINA3K prevented the intracellular calcium overload induced by H2O2. A similar protective effect was observed using a calcium chelator (BAPTA/AM). Further, SERPINA3K inhibited the phosphorylation of phospholipase C (PLC)-gamma1 induced by H2O2. Likewise, a specific PLC inhibitor showed similar protective effects on Müller cells exposed to H2O2. Furthermore, the protective effect of SERPINA3K was attenuated by a specific PLC activator (m-3M3FBS). Finally, in a binding assay, SERPINA3K displayed saturable and specific binding on Müller cells. Conclusion/Significance These results for the first time demonstrate that SERPINA3K is an endogenous serpin which protects cells from oxidative stress-induced cells death, and its protective effect is via blocking the calcium overload through the PLC pathway. The decreased retinal levels of SERPINA3K may represent a new pathogenic mechanism for the retinal Müller cell dysfunction and neuron loss in diabetes.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Cell Biology, Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jian-xing Ma
- Department of Cell Biology, Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
18
|
Initial function analysis of a novel erythroid differentiation related gene EDRF1. SCIENCE IN CHINA. SERIES C, LIFE SCIENCES 2008; 44:489-96. [PMID: 18726394 DOI: 10.1007/bf02882391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2001] [Indexed: 11/27/2022]
Abstract
Erythroid differentiation depends on the establishment of specific patterns of gene expression. Hypersensitive site 2 (HS2, serving as a major enhancer of globin genes)-binding proteins may be involved in its natural open chromosomal environment formation. Previously we prepared monoclonal antibodies against HS2-binding nuclear proteins of terminal differentiated erythroid cells. By utilizing the monoclonal antibodies, we screened lambda-gt11 human fetal liver cDNA expression library and obtained one cDNA clone, which was named erythroid differentiation related gene (EDRF1, Genbank accession number AF040247), encompassing an entire open reading frame. We investigated the expression pattern of EDRF1 by RT-PCR technique. And a clue to the function of EDRF1 has been found from confirmation of high levels of EDRF1 mRNA in differentiated K562 and human fetal liver tissue. To illuminate the function of EDRF1 in K562 cells, sense and antisense EDRF1 constructs were prepared and transfected into K562 cells. alpha-globin mRNA was down-regulated and EpoR (erythropoietin receptor) mRNA expression was increased in antisense transfected cells. Cells transfected with sense construct grew more slowly than control cells suggested by [(3)H] thimidine incorporation experiments. Suppression of K562 proliferation was accompanied by increased spontaneous hemoglobin synthesis demonstrated by spectrometry. K562 cells transfected with sense construct exhibited reduced clongenicity compared with control cells in methycellulose culture. These data provided the evidence that EDRF1 can influence globin expression and hemoglobin synthesis in K562 cells and modulated self-renewal in K562 cells.
Collapse
|
19
|
A novel benzotriazole derivative inhibits proliferation of human hepatocarcinoma cells by increasing oxidative stress concomitant mitochondrial damage. Eur J Pharmacol 2008; 584:144-52. [DOI: 10.1016/j.ejphar.2008.01.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Revised: 12/21/2007] [Accepted: 01/22/2008] [Indexed: 11/21/2022]
|
20
|
Vaqué JP, Fernández-García B, García-Sanz P, Ferrandiz N, Bretones G, Calvo F, Crespo P, Marín MC, León J. c-Myc Inhibits Ras-Mediated Differentiation of Pheochromocytoma Cells by Blocking c-Jun Up-Regulation. Mol Cancer Res 2008; 6:325-39. [DOI: 10.1158/1541-7786.mcr-07-0180] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
21
|
Sumi T, Tsuneyoshi N, Nakatsuji N, Suemori H. Apoptosis and differentiation of human embryonic stem cells induced by sustained activation of c-Myc. Oncogene 2007; 26:5564-76. [PMID: 17369859 DOI: 10.1038/sj.onc.1210353] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Embryonic stem (ES) cells are self-renewing, pluripotent cell lines, characterized by their potential to differentiate into all cell types. The proto-oncogene product c-Myc has a crucial role in the self-renewal of mouse ES (mES) cells, but its role in human ES (hES) cells is unknown. To investigate c-Myc functions in hES cells, we expressed an inducible c-Myc fused to the hormone-binding domain of the estrogen receptor (c-MycER) protein that is activated by 4-hydroxy-tamoxifen. In contrast to its role in mES cells, activation of c-MycER in hES cells induced apoptosis and differentiation into extraembryonic endoderm and trophectoderm lineages concomitant with reduced expression of the pluripotent markers Oct4 and Nanog. Neither inhibition of caspase activity nor knockdown of p53 by RNA interference impaired the induction of differentiation markers induced by c-Myc activation. In addition, differentiation induced by c-Myc activation was associated with downregulation of alpha6 integrin expression, suggesting an important role for the integrin/extracellular matrix interaction in the regulation of ES cell behavior. None of these effects occurred with deletion of the c-Myc transactivation domain, indicating that c-Myc promotes both apoptosis and differentiation in a transcriptional activity-dependent manner. Together, our results provide new insights into the c-Myc functions regulating hES cell fate.
Collapse
Affiliation(s)
- T Sumi
- Laboratory of Embryonic Stem Cell Research, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|
22
|
Yang D, Hurley LH. Structure of the biologically relevant G-quadruplex in the c-MYC promoter. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2006; 25:951-68. [PMID: 16901825 DOI: 10.1080/15257770600809913] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The nuclease hypersensitivity element III1 (NHE III1) in the c-MYC promoter controls up to 80-90% of the transcriptional activity of this gene. We have demonstrated that the guanine-rich strand of the NHE III1 forms a G-quadruplex consisting of a mixture of four biologically relevant loop isomers that function as a silencer element. NMR studies have shown that these G-quadruplexes are propeller-type parallel structures consisting of three stacked G-tetrads and three double-chain reversal loops. An NMR-derived solution structure for this quadruplex provides insight into the unusual stability of the structure. This structure is a target for small molecule inhibitors of c-MYC gene expression.
Collapse
Affiliation(s)
- Danzhou Yang
- University of Arizona, College of Pharmacy, Tucson, AZ 85721, USA
| | | |
Collapse
|
23
|
Parreño M, Vaqué JP, Casanova I, Frade P, Céspedes MV, Pavón MA, Molins A, Camacho M, Vila L, Nomdedeu JF, Mangues R, León J. Novel triiodophenol derivatives induce caspase-independent mitochondrial cell death in leukemia cells inhibited by Myc. Mol Cancer Ther 2006; 5:1166-75. [PMID: 16731748 DOI: 10.1158/1535-7163.mct-05-0257] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
2,4,6-Triiodophenol (Bobel-24, AM-24) was originally described as a nonsteroid antiinflammatory molecule. We have synthesized three derivatives of Bobel-24 (Bobel-4, Bobel-16, and Bobel-30) and tested their activities as putative antileukemic agents. We have found that Bobel-24 and Bobel-16 were dual inhibitors of cyclooxygenase and 5-lipoxygenase, whereas Bobel-4 and Bobel-30 were selective against 5-lipoxygenase. We have tested the antiproliferative activity of these compounds on a panel of cell lines derived from myeloid and lymphoid leukemias (K562, Raji, HL-60, and Molt4). The cytotoxic IC(50) in these cell lines ranged between 14 and 50 micromol/L, but it was higher for nontransformed cells such as 32D, NIH3T3, or human leukocytes. All compounds showed cytotoxic activity on all tested cell lines, accompanied by DNA synthesis inhibition and arrest in the G(0)/G(1) phase. Bobel-16, Bobel-4, and Bobel-24 induced a caspase-independent cell death in K562 and Raji cells, accompanied by chromatin condensation, cytochrome c release, and dissipation of mitochondrial membrane potential in a concentration-dependent manner and production of reactive oxygen species. As the proto-oncogene MYC is involved in mitochondrial biogenesis and survival of leukemia cells, we tested its effect on bobel activity. Bobel-24 induced down-regulation of MYC in K562 and, consistently, ectopic expression of MYC results in partial protection towards the cytotoxic effect of Bobel-24. In conclusion, Bobel derivatives induce a caspase- and Bcl-2-independent cell death in which mitochondrial permeabilization and MYC down-regulation are involved. Bobels may serve as prototypes for the development of new agents for the therapy of leukemia.
Collapse
Affiliation(s)
- Matilde Parreño
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria, Avenida Cardenal Herrera Oria, s/n 39011 Santander, Cantabria, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Peng H, Du ZW, Zhang JW. Identification and characterization of a novel zinc finger protein (HZF1) gene and its function in erythroid and megakaryocytic differentiation of K562 cells. Leukemia 2006; 20:1109-16. [PMID: 16628192 DOI: 10.1038/sj.leu.2404212] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A novel zinc finger protein (HZF1) gene was identified and characterized by screening a human bone marrow cDNA library, using a new expression sequence tag probe that contains sequences encoding zinc finger motifs. There are at least three transcripts that may result from different splicing of the pre-mRNA, but the differences among them are only involved in 5' non-translation region of HZF1 mRNA. HZF1 gene contains four exons and three introns. The putative protein consists of 670 amino-acid residues including 15 typical C2H2 and 2 C2RH zinc finger motifs. This structure characterization of HZF1 and the nuclear location of the protein suggest that HZF1 may function as a transcription factor. HZF1 mRNA expression was detected in ubiquitous tissues and various hematopoietic cell lines. Increased HZF1 mRNA expression was observed following erythroid differentiation of K562 cells induced by hemin or megakaryocytic differentiation of K562 cells induced by phorbol myristate acetate (PMA). Both of the antisense method and RNA interference assay revealed that repression of the intrinsic expression of HZF1 blocked the hemin-induced erythroid differentiation and reduced the PMA-induced megakaryocytic differentiation of K562 cells, which suggested that HZF1 play important roles in erythroid and megakaryocytic differentiation.
Collapse
Affiliation(s)
- H Peng
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | | | | |
Collapse
|
25
|
Galeano E, Nieto E, García-Pérez AI, Delgado MD, Pinilla M, Sancho P. Effects of the antitumoural dequalinium on NB4 and K562 human leukemia cell lines. Mitochondrial implication in cell death. Leuk Res 2005; 29:1201-11. [PMID: 15893819 DOI: 10.1016/j.leukres.2005.03.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Accepted: 03/15/2005] [Indexed: 10/25/2022]
Abstract
Dequalinium (DQA) is a delocalized lipophylic cation that selectively targets the mitochondria of carcinoma cells. However, the underlying mechanisms of DQA action are not yet well understood. We have studied the effects of DQA on two different leukemia cell lines: NB4, derived from acute promyelocytic leukemia, and K562, derived from chronic myeloid leukemia. We found that DQA displays differential cytotoxic activity in these cell lines. In NB4 cells, a low DQA concentration (2microM) induces a mixture of apoptosis and necrosis, whereas a high DQA concentration (20microM) induces mainly necrosis. However, K562 cell death was always by necrosis as the cells showed a resistance to apoptosis at all time-periods and DQA concentrations assayed. In both cell lines, the cell death seems to be mediated by alterations of mitochondrial function as evidenced by loss of mitochondrial transmembrane potential, O2*- accumulation and ATP depletion. The current study improves the knowledge on DQA as a novel anticancer agent with a potential application in human acute promyelocytic leukemia chemotherapy.
Collapse
Affiliation(s)
- Eva Galeano
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
26
|
Park J, Kim S, Oh C, Yoon SS, Lee D, Kim Y. Differential tyrosine phosphorylation of leukemic cells during apoptosis as a result of treatment with imatinib mesylate. Biochem Biophys Res Commun 2005; 336:942-51. [PMID: 16157305 DOI: 10.1016/j.bbrc.2005.08.201] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Accepted: 08/25/2005] [Indexed: 11/25/2022]
Abstract
Bcr-Abl fusion tyrosine kinase contributes to leukemic transformation. Imatinib mesylate inhibits Bcr-Abl tyrosine kinase, resulting in a blockage of tyrosine phosphorylation in its downstream pathways. We analyzed the alteration of tyrosine phosphorylation, on BCR/ABL+ chronic myelogenous leukemia cells, after treatment with imatinib mesylate. Data were collected using a two-dimensional gel electrophoresis followed by Western blot and mass spectrometry. The inhibition of Bcr-Abl tyrosine kinase by 2.5 microM imatinib mesylate caused both cell cycle arrest in the G0/G1 phase and increased the portion of apoptotic cells. As a result, the population of leukemic cells decreased by 30% and 70% compared to controls at 24 and 72 h, respectively. Furthermore, treatment with imatinib mesylate altered tyrosine phosphorylation of 24 protein spots as the incubation time proceeded from 0 to 24 and 72 h. Ten of the 24 protein spots are visible at all three times. Four are detectable at both the 0 and 24 h points in time. Eight were detectable only at time 0.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis
- Benzamides
- Blotting, Western
- Cell Cycle/drug effects
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Proliferation/drug effects
- Electrophoresis, Gel, Two-Dimensional
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Humans
- Imatinib Mesylate
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Phosphorylation
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Proteome/metabolism
- Pyrimidines/pharmacology
- Transcription, Genetic/drug effects
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Jungeun Park
- Division of Molecular Genomic Medicine, College of Medicine, Seoul National University, Yongon-Dong, Seoul 110-799, Republic of Korea
| | | | | | | | | | | |
Collapse
|
27
|
Maire MA, Rast C, Vasseur P. Di-(2-ethylhexyl)phthalate (DEHP) increases Bcl-2/Bax ratio and modifies c-myc expression in Syrian hamster embryo (SHE) cells. Toxicol Lett 2005; 158:237-45. [PMID: 15923092 DOI: 10.1016/j.toxlet.2005.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Revised: 04/04/2005] [Accepted: 04/05/2005] [Indexed: 11/18/2022]
Abstract
The objective of this work was to study the anti-apoptotic properties of the non-genotoxic rodent carcinogen, di(2-ethylhexyl)phthalate (DEHP) in Syrian hamster embryo (SHE) cells. We demonstrated that a 24 h pre-treatment of SHE cells with 50 microM DEHP inhibited apoptosis triggered by growth factors deprivation. The RNA expression levels of the regulator genes involved in the apoptotic pathway, bcl-2, bax and of c-myc were measured using Western blotting and RT-PCR. We showed that a 24 h treatment of SHE cells with 50 microM DEHP increased (P < 0.05) the bcl-2 expression, while c-myc expression was decreased. No effect on bax expression was observed in the range of 10-50 microM. The defective regulation of apoptosis caused by DEHP treatment could contribute to its carcinogenicity.
Collapse
Affiliation(s)
- M A Maire
- ESE, Université de Metz, Faculté des Sciences, Campus Bridoux, rue du Général Delestraint, 57070 Metz, France.
| | | | | |
Collapse
|
28
|
Halder K, Chowdhury S. Kinetic resolution of bimolecular hybridization versus intramolecular folding in nucleic acids by surface plasmon resonance: application to G-quadruplex/duplex competition in human c-myc promoter. Nucleic Acids Res 2005; 33:4466-74. [PMID: 16085756 PMCID: PMC1183106 DOI: 10.1093/nar/gki750] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The human oncogene c-myc is regulated by G-quadruplex formation within the nuclease hypersensitive element (NHE IIII) in the c-myc promoter, making the quadruplex a strong anti-cancer target. With respect to this, the competing equilibrium between intramolecular quadruplex folding and bimolecular duplex formation is poorly understood and very few techniques have addressed this problem. We present a method for simultaneously determining the kinetic constants for G-quadruplex folding/unfolding and hybridization in the presence of the complementary strand from a single reaction using an optical biosensor based on surface plasmon resonance (SPR). Using this technique, we demonstrate for the first time that quadruplex formation in the c-myc promoter is favored at low strand concentrations. Our results indicate favorable quadruplex folding (equilibrium folding constant KF of 2.09 calculated from the kinetic parameters: folding rate constant, kf = 1.65 × 10−2 s−1 and unfolding rate constant, ku = 7.90 × 10−3 s−1) in 150 mM K+. The hybridization rate constants detected concurrently gave a bimolecular association constant, ka = 1.37 × 105 M−1 s−1 and dissociation constant, kd = 4.94 × 10−5 s−1. Interestingly, in the presence of Na+ we observed that G-quadruplex folding was unfavorable (KF = 0.54). Implication of our results on the c-myc transcription activation model is discussed in light of aberrant c-myc expression observed on destabilization of the G-quadruplex.
Collapse
Affiliation(s)
| | - Shantanu Chowdhury
- To whom correspondence should be addressed. Tel: +91 11 2766 6157; Fax: +91 11 2766 7471;
| |
Collapse
|
29
|
Torrano V, Chernukhin I, Docquier F, D'Arcy V, León J, Klenova E, Delgado MD. CTCF regulates growth and erythroid differentiation of human myeloid leukemia cells. J Biol Chem 2005; 280:28152-61. [PMID: 15941718 DOI: 10.1074/jbc.m501481200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CTCF is a transcription factor and a candidate tumor suppressor that contains a DNA-binding domain composed of 11 zinc fingers. We reported previously that CTCF is differentially regulated during differentiation of human myeloid leukemia cells. In this study we aimed to investigate the role of CTCF in myeloid cell differentiation. A human cell line, K562, that can be chemically induced to differentiate into various hematopoietic lineages was chosen as a model system for this study. Several K562 cell lines with constitutive and conditional expression of CTCF have been generated. By using these model systems we demonstrated that: (i) ectopic expression of CTCF in K562 cells led to growth retardation and promotion of differentiation into the erythroid lineage; (ii) CTCF knock-down significantly inhibited differentiation of K562 cells into erythroid lineage; (iii) differentiation of K562 into the megakaryocytic lineage was not significantly affected; and (iv) down-regulation of MYC has been identified as one of the mechanisms by which CTCF promotes erythroid differentiation. Taken together our results demonstrate that CTCF is involved in the control of myeloid cell growth and differentiation.
Collapse
Affiliation(s)
- Verónica Torrano
- Grupo de Biología Molecular del Cáncer, Departamento de Biologia Molecular, Unidad de Biomedicina-CSIC, Universidad de Cantabria, 39011 Santander, Spain
| | | | | | | | | | | | | |
Collapse
|
30
|
Muñoz-Alonso MJ, Acosta JC, Richard C, Delgado MD, Sedivy J, León J. p21Cip1 and p27Kip1 Induce Distinct Cell Cycle Effects and Differentiation Programs in Myeloid Leukemia Cells. J Biol Chem 2005; 280:18120-9. [PMID: 15746092 DOI: 10.1074/jbc.m500758200] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cyclin-dependent kinase (Cdk) inhibitors p21(Cip1) and p27(Kip1) have been proposed to exert redundant functions in cell cycle progression and differentiation programs, although nonoverlapping functions have also been described. To gain further insights into the relevant mechanisms and to detect possible functional differences between both proteins, we conditionally expressed p21(Cip1) and p27(Kip1) in K562, a multipotent human leukemia cell line. Temporal ectopic expression of either p21(Cip1) or p27(Kip1) arrested proliferation, inhibited Cdk2 and Cdk4 activities, and suppressed retinoblastoma phosphorylation. However, whereas p21(Cip1) arrested cells in both G(1) and G(2) cell cycle phases, p27(Kip1) blocked the G(1)/S-phase transition. Furthermore, although both p21(Cip1) and p27(Kip1) associated with Cdk6, only p27(Kip1) significantly inhibited its activity. Most importantly, each protein promoted differentiation along a distinct pathway; p21(Cip1) triggered megakaryocytic maturation, whereas p27(Kip1) resulted in the expression of erythroid markers. Consistently, p21(Cip1) and p27(Kip1) were rapid and transiently up-regulated when K562 cells are differentiated into megakaryocytic and erythroid lineages, respectively. These findings demonstrate distinct functions of p21(Cip1) and p27(Kip1) in cell cycle regulation and differentiation and indicate that these two highly related proteins possess unique biological activities and are not functionally interchangeable.
Collapse
Affiliation(s)
- María J Muñoz-Alonso
- Grupo de Biología Molecular del Cáncer, Departamento de Biología Molecular y Unidad de Biomedicina-Consejo Superior de Investigaciones Científicas, Universidad de Cantabria, 39011 Santander, Spain
| | | | | | | | | | | |
Collapse
|
31
|
Cartwright P, McLean C, Sheppard A, Rivett D, Jones K, Dalton S. LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism. Development 2005; 132:885-96. [PMID: 15673569 DOI: 10.1242/dev.01670] [Citation(s) in RCA: 540] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Murine ES cells can be maintained as a pluripotent, self-renewing population by LIF/STAT3-dependent signaling. The downstream effectors of this pathway have not been previously defined. In this report, we identify a key target of the LIF self-renewal pathway by showing that STAT3 directly regulates the expression of the Myc transcription factor. Murine ES cells express elevated levels of Myc and following LIF withdrawal, Myc mRNA levels collapse and Myc protein becomes phosphorylated on threonine 58 (T58), triggering its GSK3beta dependent degradation. Maintained expression of stable Myc (T58A) renders self-renewal and maintenance of pluripotency independent of LIF. By contrast, expression of a dominant negative form of Myc antagonizes self-renewal and promotes differentiation. Transcriptional control by STAT3 and suppression of T58 phosphorylation are crucial for regulation of Myc activity in ES cells and therefore in promoting self-renewal. Together, our results establish a mechanism for how LIF and STAT3 regulate ES cell self-renewal and pluripotency.
Collapse
Affiliation(s)
- Peter Cartwright
- University of Georgia, Rhodes Center, 425 River Road, Athens, GA 30602-2771, USA
| | | | | | | | | | | |
Collapse
|
32
|
Vaqué JP, Navascues J, Shiio Y, Laiho M, Ajenjo N, Mauleon I, Matallanas D, Crespo P, León J. Myc antagonizes Ras-mediated growth arrest in leukemia cells through the inhibition of the Ras-ERK-p21Cip1 pathway. J Biol Chem 2004; 280:1112-22. [PMID: 15528212 DOI: 10.1074/jbc.m409503200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Even though RAS usually acts as a dominant transforming oncogene, in primary fibroblasts and some established cell lines Ras inhibits proliferation. This can explain the virtual absence of RAS mutations in some types of tumors, such as chronic myeloid leukemia (CML). We report that in the CML cell line K562 Ras induces p21Cip1 expression through the Raf-MEK-ERK pathway. Because K562 cells are deficient for p15INK4b, p16INK4a, p14ARF, and p53, this would be the main mechanism whereby Ras up-regulates p21 expression in these cells. Accordingly, we also found that Ras suppresses K562 growth by signaling through the Raf-ERK pathway. Because c-Myc and Ras cooperate in cell transformation and c-Myc is up-regulated in CML, we investigated the effect of c-Myc on Ras activity in K562 cells. c-Myc antagonized the induction of p21Cip1 mediated by oncogenic H-, K-, and N-Ras and by constitutively activated Raf and ERK2. Activation of the p21Cip1 promoter by Ras was dependent on Sp1/3 binding sites in K562. However, mutational analysis of the p21 promoter and the use of a Gal4-Sp1 chimeric protein strongly suggest that c-Myc affects Sp1 transcriptional activity but not the binding of Sp1 to the p21 promoter. c-Myc-mediated impairment of Ras activity on p21 expression required a transactivation domain, a DNA binding region, and a Max binding region. Moreover, the effect was independent of Miz1 binding to c-Myc. Consistent with its effect on p21Cip1 expression, c-Myc rescued cell growth inhibition induced by Ras. The data suggest that in particular tumor types, such as those associated with CML, c-Myc contributes to tumorigenesis by inhibiting Ras antiproliferative activity.
Collapse
Affiliation(s)
- Jose P Vaqué
- Grupo de Biología Molecular del Cáncer, Departamento de Biología Molecular, Unidad de Biomedicina del Consejo Superior de Investigaciones Cientiíficas, Facultad de Medicina, Universidad de Cantabria, 39011 Santander, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wiener Z, Ontsouka EC, Jakob S, Torgler R, Falus A, Mueller C, Brunner T. Synergistic induction of the Fas (CD95) ligand promoter by Max and NFkappaB in human non-small lung cancer cells. Exp Cell Res 2004; 299:227-35. [PMID: 15302589 DOI: 10.1016/j.yexcr.2004.05.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Revised: 04/26/2004] [Indexed: 11/28/2022]
Abstract
Fas (CD95/APO-1) ligand is a member of the Tumor Necrosis Factor family and a potent inducer of apoptosis. Fas ligand is expressed in activated T cells and represents a major cytotoxic effector mechanism by which T cells kill their target cells. Activation-induced Fas ligand expression in T cells is under the stringent control of various transcription factors, including nuclear factor kappaB (NFkappaB) and c-Myc/Max. There is accumulating evidence that Fas ligand is also expressed by various non-hematopoietic tumor cells, however, little is known about Fas ligand regulation in tumor cells. In this study, we have analyzed the regulation of the Fas ligand gene promoter induction in two non-small cell lung cancer cell lines, with a major focus on the role of the c-Myc/Max transcription factor. Our results revealed that inhibition of c-Myc/Max did not substantially reduce basal levels of Fas ligand promoter activity, nor did overexpression of c-Myc significantly induce promoter activity. In contrast, we observed that overexpression of Max resulted in a marked increase in basal promoter activity and synergistically enhanced phorbolester- and doxorubicin-induced NFkappaB-mediated Fas ligand promoter activity. These results were confirmed by analyzing endogenous Fas ligand transcription. We conclude that high levels of Max and stress-induced NFkappaB activation may result in elevated expression of Fas ligand in human lung cancer cells and possibly contribute to Fas ligand-associated immune escape mechanisms.
Collapse
MESH Headings
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors
- Basic-Leucine Zipper Transcription Factors
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Cell Line, Tumor
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Doxorubicin/pharmacology
- Fas Ligand Protein
- Gene Expression Regulation, Neoplastic/genetics
- Genes, Regulator/genetics
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Membrane Glycoproteins/genetics
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Phorbol Esters/pharmacology
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Proteins c-myc/genetics
- Stress, Physiological/genetics
- Stress, Physiological/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic/genetics
- Tumor Escape/genetics
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Zoltan Wiener
- Division of Immunopathology, Institute of Pathology, University of Berne, Berne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
34
|
Li CY, Zhan YQ, Xu CW, Xu WX, Wang SY, Lv J, Zhou Y, Yue PB, Chen B, Yang XM. EDAG regulates the proliferation and differentiation of hematopoietic cells and resists cell apoptosis through the activation of nuclear factor-κB. Cell Death Differ 2004; 11:1299-308. [PMID: 15332117 DOI: 10.1038/sj.cdd.4401490] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Erythroid differentiation-associated gene (EDAG) is considered to be a human hematopoiesis-specific gene. Here, we reported that downregulation of EDAG protein in K562 cells resulted in inhibition of growth and colony formation, and enhancement of sensitivity to erythroid differentiation induced by hemin. Overexpression of EDAG in HL-60 cells significantly blocked the expression of the monocyte/macrophage differentiation marker CD11b after pentahydroxytiglia myristate acetate induction. Moreover, overexpression of EDAG in pro-B Ba/F3 cells prolonged survival and increased the expression of c-Myc, Bcl-2 and Bcl-xL in the absence of interleukin-3 (IL-3). Furthermore, we showed that EDAG enhanced the transcriptional activity of nuclear factor-kappa B (NF-kappa B), and high DNA-binding activity of NF-kappa B was sustained in Ba/F3 EDAG cells after IL-3 was withdrawn. Inhibition of NF-kappa B activity resulted in promoting Ba/F3 EDAG cells death. These results suggest that EDAG regulates the proliferation and differentiation of hematopoietic cells and resists cell apoptosis through the activation of NF-kappa B.
Collapse
Affiliation(s)
- C Y Li
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Petrs-Silva H, de Freitas FG, Linden R, Chiarini LB. Early nuclear exclusion of the transcription factor max is associated with retinal ganglion cell death independent of caspase activity. J Cell Physiol 2003; 198:179-87. [PMID: 14603520 DOI: 10.1002/jcp.10404] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We examined the behavior of the transcription factor Max during retrograde neuronal degeneration of retinal ganglion cells. Using immunohistochemistry, we found a progressive redistribution of full-length Max from the nucleus to the cytoplasm and dendrites of the ganglion cells following axon damage. Then, the axotomized cells lose all their content of Max, while undergoing nuclear pyknosis and apoptotic cell death. After treatment of retinal explants with either anisomycin or thapsigargin, the rate of nuclear exclusion of Max accompanied the rate of cell death as modulated by either drug. Treatment with a pan-caspase inhibitor abolished both TUNEL staining and immunoreactivity for activated caspase-3, but did not affect the subcellular redistribution of Max immunoreactivity after axotomy. The data show that nuclear exclusion of the transcription factor Max is an early event, which precedes and is independent of the activation of caspases, during apoptotic cell death in the central nervous system.
Collapse
Affiliation(s)
- Hilda Petrs-Silva
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
36
|
Melkoumian ZK, Martirosyan AR, Strobl JS. Myc protein is differentially sensitive to quinidine in tumor versus immortalized breast epithelial cell lines. Int J Cancer 2002; 102:60-9. [PMID: 12353235 DOI: 10.1002/ijc.10648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Quinidine regulates growth and differentiation in human breast tumor cells, but the immortalized mammary epithelial MCF-10A cell line is insensitive to quinidine. We found that a morphologically similar differentiation response was evoked by quinidine and c-myc antisense oligonucleotides in MCF-7 cells and this prompted us to investigate the actions of quinidine on c-myc gene expression. Myc protein levels were suppressed in human breast tumor cell lines, but not in MCF-10A cells, an observation that supports the hypothesis that suppression of c-myc gene expression is involved in the preferential growth and differentiation response of breast tumor cells to quinidine. Quinidine reduced c-myc mRNA levels in MCF-7 cells. Acute induction of c-myc mRNA by estradiol, as well as the c-myc response to sub-cultivation in fresh serum and H-ras driven elevations in c-myc mRNA were depressed by 50-60% in the presence of quinidine. Quinidine decreased c-myc promoter activity in MCF-7 cells in a transient reporter gene assay and a 168 bp region of human c-myc promoter (-100 to +68 with respect to the P1 promoter) was sufficient to confer responsiveness to quinidine. Quinidine is a potential lead compound for developing pharmacological agents to regulate Myc. In addition, the study of quinidine-regulated events is a promising approach to unravel differentiation control pathways that become disrupted in breast cancer.
Collapse
Affiliation(s)
- Zaroui K Melkoumian
- Department of Biochemistry and Molecular Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | | | | |
Collapse
|
37
|
Siddiqui-Jain A, Grand CL, Bearss DJ, Hurley LH. Direct evidence for a G-quadruplex in a promoter region and its targeting with a small molecule to repress c-MYC transcription. Proc Natl Acad Sci U S A 2002; 99:11593-8. [PMID: 12195017 PMCID: PMC129314 DOI: 10.1073/pnas.182256799] [Citation(s) in RCA: 1840] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The nuclease hypersensitivity element III(1) upstream of the P1 promoter of c-MYC controls 85-90% of the transcriptional activation of this gene. We have demonstrated that the purine-rich strand of the DNA in this region can form two different intramolecular G-quadruplex structures, only one of which seems to be biologically relevant. This biologically relevant structure is the kinetically favored chair-form G-quadruplex, which is destabilized when mutated with a single G --> A transition, resulting in a 3-fold increase in basal transcriptional activity of the c-MYC promoter. The cationic porphyrin TMPyP4, which has been shown to stabilize this G-quadruplex structure, is able to suppress further c-MYC transcriptional activation. These results provide compelling evidence that a specific G-quadruplex structure formed in the c-MYC promoter region functions as a transcriptional repressor element. Furthermore, we establish the principle that c-MYC transcription can be controlled by ligand-mediated G-quadruplex stabilization.
Collapse
|
38
|
Adachi S, Obaya AJ, Han Z, Ramos-Desimone N, Wyche JH, Sedivy JM. c-Myc is necessary for DNA damage-induced apoptosis in the G(2) phase of the cell cycle. Mol Cell Biol 2001; 21:4929-37. [PMID: 11438650 PMCID: PMC87219 DOI: 10.1128/mcb.21.15.4929-4937.2001] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The c-myc proto-oncogene encodes a transcription factor that participates in the regulation of cellular proliferation, differentiation, and apoptosis. Ectopic overexpression of c-Myc has been shown to sensitize cells to apoptosis. We report here that cells lacking c-Myc activity due to disruption of the c-myc gene by targeted homologous recombination are defective in DNA damage-initiated apoptosis in the G(2) phase of the cell cycle. The downstream effector of c-Myc is cyclin A, whose ectopic expression in c-myc(-/-) cells rescues the apoptosis defect. The kinetics of the G(2) response indicate that the induction of cyclin A and the concomitant activation of Cdk2 represent an early step during commitment to apoptosis. In contrast, expression of cyclins E and D1 does not rescue the apoptosis defect, and apoptotic processes in G(1) phase are not affected in c-myc(-/-) cells. These observations link DNA damage-induced apoptosis with cell cycle progression and implicate c-Myc in the functioning of a subset of these pathways.
Collapse
Affiliation(s)
- S Adachi
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912, USA
| | | | | | | | | | | |
Collapse
|
39
|
Lin KI, Lin Y, Calame K. Repression of c-myc is necessary but not sufficient for terminal differentiation of B lymphocytes in vitro. Mol Cell Biol 2000; 20:8684-95. [PMID: 11073970 PMCID: PMC86481 DOI: 10.1128/mcb.20.23.8684-8695.2000] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The importance of c-myc as a target of the Blimp-1 repressor has been studied in BCL-1 cells, in which Blimp-1 is sufficient to trigger terminal B-cell differentiation. Our data show that Blimp-1-dependent repression of c-myc is required for BCL-1 differentiation, since constitutive expression of c-Myc blocked differentiation. Furthermore, ectopic expression of cyclin E mimicked the effects of c-Myc on both proliferation and differentiation, indicating that the ability of c-Myc to drive proliferation is responsible for blocking BCL-1 differentiation. However, inhibition of c-Myc by a dominant negative form was not sufficient to drive BCL-1 differentiation. Thus, during Blimp-1-dependent plasma cell differentiation, repression of c-myc is necessary but not sufficient, demonstrating the existence of additional Blimp-1 target genes.
Collapse
Affiliation(s)
- K I Lin
- Department of Microbiology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | |
Collapse
|
40
|
Ceballos E, Delgado MD, Gutierrez P, Richard C, Müller D, Eilers M, Ehinger M, Gullberg U, León J. c-Myc antagonizes the effect of p53 on apoptosis and p21WAF1 transactivation in K562 leukemia cells. Oncogene 2000; 19:2194-204. [PMID: 10822369 DOI: 10.1038/sj.onc.1203541] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
c-myc protooncogene positively regulates cell proliferation and overexpression of c-myc is found in many solid tumors and leukemias. In the present study we used the K562 human myeloid leukemia cell line as a model to study the functional interaction between c-Myc and p53. Using two different methods, we generated K562 transfectant cell lines with conditional expression of either c-Myc or p53. The cells expressed the p53Vall35 mutant, which adopts a wild-type conformation at 32 degrees C, while c-Myc induction was achieved with a zinc-inducible expression vector. We found that p53 in wild-type conformation induces growth arrest and apoptosis of K562. Expression of c-Myc significantly attenuated apoptosis and impaired the transcriptional activity of p53 on p21WAF1, Bax and cytomegalovirus promoters. The impairment of p21WAF1 transactivation by c-Myc was confirmed by transfection of a c-Myc-estrogen receptor fusion protein and by induction of c-myc by zinc in transfected cells. Also, p53-mediated up-regulation of p21WAF1 mRNA protein were significantly reduced by c-Myc, while Bax levels were unaffected. Consistently, c-Myc increased cyclin-dependent kinase 2 activity in K562 cells expressing p53 in wild-type conformation. These results suggest that c-Myc overexpression may antagonize the pro-apoptotic function of p53, thus providing a molecular mechanism for the frequently observed deregulation of c-myc in human cancer.
Collapse
Affiliation(s)
- E Ceballos
- Departamento de Biologia Molecular, Unidad Asociada al Centro de Investigaciones Biológicas, Universidad de Cantabria, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Delgado MD, Chernukhin IV, Bigas A, Klenova EM, León J. Differential expression and phosphorylation of CTCF, a c-myc transcriptional regulator, during differentiation of human myeloid cells. FEBS Lett 1999; 444:5-10. [PMID: 10037138 DOI: 10.1016/s0014-5793(99)00013-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
CTCF is a transcriptional repressor of the c-myc gene. Although CTCF has been characterized in some detail, there is very little information about the regulation of CTCF activity. Therefore we investigated CTCF expression and phosphorylation during induced differentiation of human myeloid leukemia cells. We found that: (i) both CTCF mRNA and protein are down-regulated during terminal differentiation in most cell lines tested; (ii) CTCF down-regulation is retarded and less pronounced than that of c-myc; (iii) CTCF protein is differentially phosphorylated and the phosphorylation profiles depend on the differentiation pathway. We concluded that CTCF expression and activity is controlled at transcriptional and post-transcriptional levels.
Collapse
Affiliation(s)
- M D Delgado
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | | | | | | | | |
Collapse
|
42
|
Delgado MD, Gutiérrez P, Richard C, Cuadrado MA, Moreau-Gachelin F, León J. Spi-1/PU.1 proto-oncogene induces opposite effects on monocytic and erythroid differentiation of K562 cells. Biochem Biophys Res Commun 1998; 252:383-91. [PMID: 9826539 DOI: 10.1006/bbrc.1998.9587] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Spi-1/PU.1 is a hematopoietic transcription factor of the Ets family. To analyze the effects of ectopic expression of spi-1 on the proliferation/differentiation of human myeloid leukemia cells, K562 cells were stably transfected with a spi-1 expression vector. The transfected cell lines expressed elevated levels of spi-1 mRNA and protein and high Spi-1-DNA binding activity. The spi-1 transfected cells showed reduced growth rates and reduced clonogenic cell growth. When the erythroid and monocytic differentiation markers were analyzed, spi-1 overexpression resulted in opposite effects: erythroid differentiation was significantly inhibited in spi-1 transfectants, while spi-1 overexpression increased the monocytic differentiation of cells. These results indicate a differential role of Spi-1 on the differentiation of human myeloid leukemia cells.
Collapse
Affiliation(s)
- M D Delgado
- Grupo de Biología Molecular del Cáncer, Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria, Santander, 39011, Spain
| | | | | | | | | | | |
Collapse
|
43
|
Facchini LM, Penn LZ. The molecular role of Myc in growth and transformation: recent discoveries lead to new insights. FASEB J 1998. [DOI: 10.1096/fasebj.12.9.633] [Citation(s) in RCA: 278] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Linda M. Facchini
- Ontario Cancer Institute/Princess Margaret HospitalDivision of Cell and Molecular BiologyDepartment of Medical BiophysicsUniversity of Toronto Toronto M5G 2M9 Canada
| | - Linda Z. Penn
- Ontario Cancer Institute/Princess Margaret HospitalDivision of Cell and Molecular BiologyDepartment of Medical BiophysicsUniversity of Toronto Toronto M5G 2M9 Canada
| |
Collapse
|
44
|
Gutiérrez P, Delgado MD, Richard C, Moreau-Gachelin F, León J. Interferon induces up-regulation of Spi-1/PU.1 in human leukemia K562 cells. Biochem Biophys Res Commun 1997; 240:862-8. [PMID: 9398659 DOI: 10.1006/bbrc.1997.7760] [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
The human K562 cell line is derived from a chronic myelogenous leukemia in blastic crisis. Treatment of K562 cells with interferons alpha, beta or gamma resulted in inhibition of cell proliferation. Spi-1/PU.1 is a transcription factor of the Ets family which is required for normal hematopoyesis. We have found that spi-1 mRNA and protein as well as Spi-1-DNA binding activity increase after exposure of K562 cells to interferons. The increase in spi-1 expression ranged from 4- to 8-fold with the different interferons. K562 cells can be differentiated in vitro towards erythroid cells or monocyte-macrophage cells. Interestingly, the regulation of spi-1 by interferon-alpha depended on the differentiated phenotype of K562 cells: interferon-alpha failed to induce spi-1 in erythroid differentiated cells, whereas it induced spi-1 in monocyte-macrophage differentiated cells. The results suggest a role for Spi-1 in the cytostatic response to interferons.
Collapse
MESH Headings
- Animals
- Blast Crisis
- Blotting, Northern
- Blotting, Western
- COS Cells
- Cell Differentiation
- Cell Division
- DNA, Neoplasm/metabolism
- Gene Expression Regulation, Neoplastic
- Hematopoiesis
- Humans
- Interferons/pharmacology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Nuclear Proteins/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Messenger/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transfection
- Tumor Cells, Cultured
- Up-Regulation
Collapse
Affiliation(s)
- P Gutiérrez
- Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | | | | | | | | |
Collapse
|
45
|
Abstract
The epidermis contains two types of proliferative keratinocyte: stem cells, with unlimited self-renewal capacity, and transit amplifying cells, those daughters of stem cells that are destined to withdraw from the cell cycle and terminally differentiate after a few rounds of division. In a search for factors that regulate exit from the stem cell compartment, we constitutively expressed c-Myc in primary human keratinocytes by use of wild-type and steroid-activatable constructs. In contrast to its role in other cell types, activation of c-Myc in keratinocytes caused a progressive reduction in growth rate, without inducing apoptosis, and a marked stimulation of terminal differentiation. Keratinocytes can be enriched for stem or transit amplifying cells on the basis of beta1 integrin expression and by use of this method to fractionate cells prior to c-Myc activation, we found that c-Myc acted selectively on stem cells, driving them into the transit amplifying compartment. As a result, activation of c-Myc in epidermis reconstituted on a dermal equivalent led to premature execution of the differentiation program. The transcriptional regulatory domain of c-Myc was required for these effects because a deletion within that domain acted as a dominant-negative mutation. Our results reveal a novel biological role for c-Myc and provide new insights into the mechanism regulating epidermal stem cell fate.
Collapse
Affiliation(s)
- A Gandarillas
- Keratinocyte Laboratory, Imperial Cancer Research Fund (ICRF), London, WC2A 3PX, UK
| | | |
Collapse
|