1
|
Đukić A, Lulić L, Thomas M, Skelin J, Bennett Saidu NE, Grce M, Banks L, Tomaić V. HPV Oncoproteins and the Ubiquitin Proteasome System: A Signature of Malignancy? Pathogens 2020; 9:pathogens9020133. [PMID: 32085533 PMCID: PMC7168213 DOI: 10.3390/pathogens9020133] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/22/2022] Open
Abstract
Human papillomavirus (HPV) E6 and E7 oncoproteins are critical for development and maintenance of the malignant phenotype in HPV-induced cancers. These two viral oncoproteins interfere with a plethora of cellular pathways, including the regulation of cell cycle and the control of apoptosis, which are critical in maintaining normal cellular functions. E6 and E7 bind directly with certain components of the Ubiquitin Proteasome System (UPS), enabling them to manipulate a number of important cellular pathways. These activities are the means by which HPV establishes an environment supporting the normal viral life cycle, however in some instances they can also lead to the development of malignancy. In this review, we have discussed how E6 and E7 oncoproteins from alpha and beta HPV types interact with the components of the UPS, and how this interplay contributes to the development of cancer.
Collapse
Affiliation(s)
- Anamaria Đukić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (A.Đ.); (L.L.); (J.S.); (N.E.B.S.); (M.G.)
| | - Lucija Lulić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (A.Đ.); (L.L.); (J.S.); (N.E.B.S.); (M.G.)
| | - Miranda Thomas
- International Centre for Genetic Engineering and Biotechnology, AREA Science Park, Padriciano 99, I-34149 Trieste, Italy; (M.T.); (L.B.)
| | - Josipa Skelin
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (A.Đ.); (L.L.); (J.S.); (N.E.B.S.); (M.G.)
| | - Nathaniel Edward Bennett Saidu
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (A.Đ.); (L.L.); (J.S.); (N.E.B.S.); (M.G.)
| | - Magdalena Grce
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (A.Đ.); (L.L.); (J.S.); (N.E.B.S.); (M.G.)
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, AREA Science Park, Padriciano 99, I-34149 Trieste, Italy; (M.T.); (L.B.)
| | - Vjekoslav Tomaić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia; (A.Đ.); (L.L.); (J.S.); (N.E.B.S.); (M.G.)
- Correspondence: ; Tel.: +385-1-4561110; Fax: +385-1-4561010
| |
Collapse
|
2
|
Suppression of BCL6 function by HDAC inhibitor mediated acetylation and chromatin modification enhances BET inhibitor effects in B-cell lymphoma cells. Sci Rep 2019; 9:16495. [PMID: 31712669 PMCID: PMC6848194 DOI: 10.1038/s41598-019-52714-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple genetic aberrations in the regulation of BCL6, including in acetyltransferase genes, occur in clinically aggressive B-cell lymphomas and lead to higher expression levels and activity of this transcriptional repressor. BCL6 is, therefore, an attractive target for therapy in aggressive lymphomas. In this study romidepsin, a potent histone deacetylase inhibitor (HDACi), induced apoptosis and cell cycle arrest in Burkitt and diffuse large B-cell lymphoma cell lines, which are model cells for studying the mechanism of action of BCL6. Romidepsin caused BCL6 acetylation at early timepoints inhibiting its function, while at later timepoints BCL6 expression was reduced and target gene expression increased due to chromatin modification. MYC contributes to poor prognosis in aggressive lymphoma. MYC function is reduced by inhibition of chromatin readers of the bromodomain and extra-terminal repeat (BET) family, which includes BRD4. The novel combination of romidepsin and JQ1, a BRD4 inhibitor was investigated and showed synergy. Collectively we suggest that the combination of HDACi and BRD4i should be pursued in further pre-clinical testing.
Collapse
|
3
|
McConnell MJ, Durand L, Langley E, Coste-Sarguet L, Zelent A, Chomienne C, Kouzarides T, Licht JD, Guidez F. Post transcriptional control of the epigenetic stem cell regulator PLZF by sirtuin and HDAC deacetylases. Epigenetics Chromatin 2015; 8:38. [PMID: 26405459 PMCID: PMC4581162 DOI: 10.1186/s13072-015-0030-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/14/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The transcriptional repressor promyelocytic leukemia zinc finger protein (PLZF) is critical for the regulation of normal stem cells maintenance by establishing specific epigenetic landscape. We have previously shown that CBP/p300 acetyltransferase induces PLZF acetylation in order to increase its deoxynucleotidic acid (DNA) binding activity and to enhance its epigenetic function (repression of PLZF target genes). However, how PLZF is inactivated is not yet understood. RESULTS In this study, we demonstrate that PLZF is deacetylated by both histone deacetylase 3 and the NAD+ dependent deacetylase silent mating type information regulation 2 homolog 1 (SIRT1). Unlike other PLZF-interacting deacetylases, these two proteins interact with the zinc finger domain of PLZF, where the activating CBP/p300 acetylation site was previously described, inducing deacetylation of lysines 647/650/653. Overexpression of histone deacetylase 3 (HDAC3) and SIRT1 is associated with loss of PLZF DNA binding activity and decreases PLZF transcriptional repression. As a result, the chromatin status of the promoters of PLZF target genes, involved in oncogenesis, shift from a heterochromatin to an open euchromatin environment leading to gene expression even in the presence of PLZF. CONCLUSIONS Consequently, SIRT1 and HDAC3 mediated-PLZF deacetylation provides for rapid control and fine-tuning of PLZF activity through post-transcriptional modification to regulate gene expression and cellular homeostasis.
Collapse
Affiliation(s)
- Melanie J. McConnell
- />Malaghan Institute for Medical Research, P.O. Box 7060, Wellington, New Zealand
- />Division of Hematology/Oncology, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Laetitia Durand
- />INSERM UMRS-1131, Institut universitaire d’Hématologie, Université Paris Diderot, 1 avenue Claude Vellefaux, hôpital Saint-Louis, 75010 Paris, France
| | - Emma Langley
- />Wellcome Institute/Cancer Research UK, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QR UK
- />Biogen Idec, San Diego, CA 92122 USA
| | - Lise Coste-Sarguet
- />INSERM UMRS-1131, Institut universitaire d’Hématologie, Université Paris Diderot, 1 avenue Claude Vellefaux, hôpital Saint-Louis, 75010 Paris, France
| | - Arthur Zelent
- />Division of Hemato-oncology, Miller School of Medicine, Miami, FL 33136 USA
| | - Christine Chomienne
- />INSERM UMRS-1131, Institut universitaire d’Hématologie, Université Paris Diderot, 1 avenue Claude Vellefaux, hôpital Saint-Louis, 75010 Paris, France
| | - Tony Kouzarides
- />Wellcome Institute/Cancer Research UK, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QR UK
| | - Jonathan D. Licht
- />Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 USA
- />Division of Hematology/Oncology, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Fabien Guidez
- />INSERM UMRS-1131, Institut universitaire d’Hématologie, Université Paris Diderot, 1 avenue Claude Vellefaux, hôpital Saint-Louis, 75010 Paris, France
| |
Collapse
|
4
|
Scofield MD, Korutla L, Jackson TG, Kalivas PW, Mackler SA. Nucleus Accumbens 1, a Pox virus and Zinc finger/Bric-a-brac Tramtrack Broad protein binds to TAR DNA-binding protein 43 and has a potential role in Amyotrophic Lateral Sclerosis. Neuroscience 2012; 227:44-54. [PMID: 23022214 DOI: 10.1016/j.neuroscience.2012.09.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 09/18/2012] [Accepted: 09/19/2012] [Indexed: 12/12/2022]
Abstract
Protein degradation is a critical component of cellular maintenance. The intracellular translocation and targeting of the Ubiquitin Proteasome System (UPS) differentially coordinates a protein's half-life and thereby its function. Nucleus Accumbens 1 (NAC1), a member of the Pox virus and Zinc finger/Bric-a-brac Tramtrack Broad complex (POZ/BTB) family of proteins, participates in the coordinated proteolysis of synaptic proteins by mediating recruitment of the UPS to dendritic spines. Here we report a novel interaction between NAC1 and TAR DNA-binding protein 43 (TDP-43), a protein identified as the primary component of ubiquitinated protein aggregates found in patients with Amyotrophic Lateral Sclerosis (ALS). In vitro translated full-length TDP-43 associated with both the POZ/BTB domain and the non-POZ/BTB domain of NAC1 in GST pulldown assays. Other POZ/BTB proteins (including zinc finger POZ/BTB proteins and atypical POZ/BTB proteins) showed weak interactions with TDP-43. In addition, NAC1 and TDP-43 were present in the same immunocomplexes in different regions of mouse brain and spinal cord. In primary spinal cord cultures, TDP-43 expression was mainly nuclear, whereas NAC1 was both nuclear and cytoplasmic. In order to mimic ALS-like toxicity in the spinal cord culture system, we elevated extracellular glutamate levels resulting in the selective loss of motor neurons. Using this model, it was found that glutamate toxicity elicited a dose-dependent translocation of TDP-43 out of the nucleus of cholinergic neurons and increased the co-localization of NAC1 and TDP-43. These findings suggest that NAC1 may function to link TDP-43 to the proteasome; thereby, facilitating the post-translational modifications of TDP-43 that lead to the development of ALS.
Collapse
Affiliation(s)
- M D Scofield
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA.
| | | | | | | | | |
Collapse
|
5
|
Calderaro J, Moroch J, Pierron G, Pedeutour F, Grison C, Maillé P, Soyeux P, de la Taille A, Couturier J, Vieillefond A, Rousselet MC, Delattre O, Allory Y. SMARCB1/INI1 inactivation in renal medullary carcinoma. Histopathology 2012; 61:428-35. [DOI: 10.1111/j.1365-2559.2012.04228.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
6
|
Bonadies N, Foster SD, Chan WI, Kvinlaug BT, Spensberger D, Dawson MA, Spooncer E, Whetton AD, Bannister AJ, Huntly BJ, Göttgens B. Genome-wide analysis of transcriptional reprogramming in mouse models of acute myeloid leukaemia. PLoS One 2011; 6:e16330. [PMID: 21297973 PMCID: PMC3030562 DOI: 10.1371/journal.pone.0016330] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 12/12/2010] [Indexed: 11/27/2022] Open
Abstract
Acute leukaemias are commonly caused by mutations that corrupt the transcriptional circuitry of haematopoietic stem/progenitor cells. However, the mechanisms underlying large-scale transcriptional reprogramming remain largely unknown. Here we investigated transcriptional reprogramming at genome-scale in mouse retroviral transplant models of acute myeloid leukaemia (AML) using both gene-expression profiling and ChIP-sequencing. We identified several thousand candidate regulatory regions with altered levels of histone acetylation that were characterised by differential distribution of consensus motifs for key haematopoietic transcription factors including Gata2, Gfi1 and Sfpi1/Pu.1. In particular, downregulation of Gata2 expression was mirrored by abundant GATA motifs in regions of reduced histone acetylation suggesting an important role in leukaemogenic transcriptional reprogramming. Forced re-expression of Gata2 was not compatible with sustained growth of leukaemic cells thus suggesting a previously unrecognised role for Gata2 in downregulation during the development of AML. Additionally, large scale human AML datasets revealed significantly higher expression of GATA2 in CD34+ cells from healthy controls compared with AML blast cells. The integrated genome-scale analysis applied in this study represents a valuable and widely applicable approach to study the transcriptional control of both normal and aberrant haematopoiesis and to identify critical factors responsible for transcriptional reprogramming in human cancer.
Collapse
Affiliation(s)
- Nicolas Bonadies
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Samuel D. Foster
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Wai-In Chan
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Brynn T. Kvinlaug
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Dominik Spensberger
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Mark A. Dawson
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Elaine Spooncer
- School of Cancer and Imaging Sciences, University of Manchester, Manchester, United Kingdom
| | - Anthony D. Whetton
- School of Cancer and Imaging Sciences, University of Manchester, Manchester, United Kingdom
| | - Andrew J. Bannister
- Gurdon Institute and Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Brian J. Huntly
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
7
|
Abstract
BCL6 is a transcription factor that has essential B-cell and T-cell roles in normal antibody responses. It is involved in chromosomal translocations in diffuse large B-cell lymphoma (DBCL; including primary mediastinal B-cell lymphoma) and nodular lymphocyte predominant Hodgkin lymphoma, and is expressed in follicular lymphoma and Burkitt's lymphoma. The neoplastic T-cells of angioimmunoblastic T-cell lymphoma also express BCL6. BCL6 prevents terminal B-cell differentiation largely through repression of PRDM1. In the "cell of origin" classification of DLBCL BCL6 is associated with the germinal centre subtype, which carries a good response to modern treatments. More recently, specific BCL6 antagonists, including small molecule inhibitors, have been developed. These antagonists have demonstrated that DLBCL cells, in which BCL6 is transcriptionally active, are dependent on this gene for survival. BCL6 antagonists are active against primary DLBCL and may find future application in the treatment of lymphomas.
Collapse
Affiliation(s)
- Simon D Wagner
- Department of Cancer Studies and Molecular Medicine and MRC Toxicology Unit, University of Leicester, Lancaster Road, Leicester, UK.
| | | | | |
Collapse
|
8
|
Abstract
Although the majority of children with acute lymphoblastic leukemia (ALL) can be cured with combination chemotherapy, the challenge remains to salvage patients with resistant disease and to reduce treatment related toxicity. To meet this challenge, it will be essential to incorporate new agents targeting the biological Achilles Heels of this cancer more rapidly into currently available treatment regimen. Here we review the principles of current ALL therapy, recent advances in understanding ALL biology and discuss a selection of promising areas for drug development that may take advantage of the underlying leukemia biology. We focus particularly on strategies to interfere with common effector mechanisms that can be trigged by different individual oncogenic lesions and on new agents from drug development programs in adult oncology, as such agents will come with better chances for sustainable commercial development.
Collapse
|
9
|
Abstract
Acute myeloid leukemia (AML) is characterized by molecular heterogeneity that is not fully reflected in the current classification system. Recent insights point toward a significant role of aberrant DNA methylation in leukemogenesis. Therefore, we investigated the prognostic impact of DNA methylation in AML. To screen for promoter methylation in AML we applied a combination of base-specific cleavage biochemistry and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS), a powerful methodology allowing for quantitatively investigating DNA methylation status in a large series of both promoter regions and leukemia samples. We analyzed 92 genomic regions in 182 patient samples, correlated findings with clinical and molecular data, and validated the results in an independent cohort of 74 AML samples. Using this approach, we were able to identify novel leukemia subgroups based on distinct DNA methylation patterns. Furthermore, we defined a methylation-based outcome predictor for patient survival (P < .01) that in multivariable analysis provided independent prognostic information (hazard ratio, 1.52; 95% CI, 1.06-2.16). Here, we report the first large-scale methylation-based outcome predictor in AML, and thereby our findings support the use of genomic methylation markers for improved molecular classification and prognostication in adult AML.
Collapse
|
10
|
|
11
|
Fiskus W, Buckley K, Rao R, Mandawat A, Yang Y, Joshi R, Wang Y, Balusu R, Chen J, Koul S, Joshi A, Upadhyay S, Atadja P, Bhalla KN. Panobinostat treatment depletes EZH2 and DNMT1 levels and enhances decitabine mediated de-repression of JunB and loss of survival of human acute leukemia cells. Cancer Biol Ther 2009; 8:939-50. [PMID: 19279403 DOI: 10.4161/cbt.8.10.8213] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The PRC2 complex protein EZH2 is a histone methyltransferase that is known to bind and recruit DNMT1 to the DNA to modulate DNA methylation. Here, we determined that the pan-HDAC inhibitor panobinostat (LBH589) treatment depletes DNMT1 and EZH2 protein levels, disrupts the interaction of DNMT1 with EZH2, as well as de-represses JunB in human acute leukemia cells. Similar to treatment with the hsp90 inhibitor 17-DMAG, treatment with panobinostat also inhibited the chaperone association of heat shock protein 90 with DNMT1 and EZH2, which promoted the proteasomal degradation of DNMT1 and EZH2. Unlike treatment with the DNA methyltransferase inhibitor decitabine, which demethylates JunB promoter DNA, panobinostat treatment mediated chromatin alterations in the JunB promoter. Combined treatment with panobinostat and decitabine caused greater attenuation of DNMT1 and EZH2 levels than either agent alone, which was accompanied by more JunB de-repression and loss of clonogenic survival of K562 cells. Co-treatment with panobinostat and decitabine also caused more loss of viability of primary AML but not normal CD34(+) bone marrow progenitor cells. Collectively, these findings indicate that co-treatment with panobinostat and decitabine targets multiple epigenetic mechanisms to de-repress JunB and exerts antileukemia activity against human acute myeloid leukemia cells.
Collapse
Affiliation(s)
- Warren Fiskus
- MCG Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
NAC1, a POZ/BTB protein that functions as a corepressor. Neurochem Int 2008; 54:245-52. [PMID: 19121354 DOI: 10.1016/j.neuint.2008.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 11/25/2008] [Accepted: 12/02/2008] [Indexed: 11/20/2022]
Abstract
We now demonstrate that NAC1 acts as a corepressor for other POZ/BTB proteins. NAC1 is a POZ/BTB motif containing transcriptional repressor protein. In a mammalian two hybrid assay in neuronal (N2A) cells and non-neuronal (HEK 293T) cells, VP16 activation domain tagged NAC1 resulted in significant reversal of transcriptional inhibition with the Gal4-ZID, Gal4-BCL6, Gal4-ZF5, and kelch proteins Gal4-MAYVEN and Gal4-NRP/B fusion proteins. We also observed similar results with another corepressor, BCoR Gal4 fusion protein. NAC1 potentiated ZF5 mediated repression in Gal4-DBD fusion transient assays. GST pulldown assays further confirmed protein-protein interactions between these proteins and NAC1. Both the NAC1 isoforms demonstrated selective interaction through the POZ/BTB domain but not with the non-POZ/BTB region. Endogenous NAC1 and BCL6 physically associated in CNS regions. Strikingly, NAC1 did not interact with the pro-myelocytic leukemia zinc finger protein (PLZF), another POZ/BTB protein that is not found in the adult brain. Therefore, we conclude that NAC1 functions as a corepressor for POZ/BTB proteins expressed in the mature CNS.
Collapse
|
13
|
The MMSET protein is a histone methyltransferase with characteristics of a transcriptional corepressor. Blood 2007; 111:3145-54. [PMID: 18156491 DOI: 10.1182/blood-2007-06-092122] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
MMSET, identified by its fusion to the IgH locus in t(4;14)-associated multiple myeloma, possesses domains found within chromatin regulators, including the SET domain. MMSET protein is overexpressed and highly associated with chromatin in myeloma cell lines carrying t(4;14). MMSET possesses methyltransferase activity for core histone H3 lysine 4 and histone 4 lysine 20, whereas MMSET made in cells only modified H4. Segments of MMSET fused to the Gal4 DNA binding domain repressed transcription of a chromatin-embedded Gal4 reporter gene. MMSET-mediated repression was associated with increased H4K20 methylation gene and loss of histone acetylation. Consistent with this repressive activity, MMSET could form a complex with HDAC1 and HDAC2, mSin3a, and the histone demethylase LSD1, suggesting that it is a component of corepressor complexes. Furthermore, MMSET coexpression enhances HDAC1- and HDAC2-mediated repression in transcriptional reporter assays. Finally, shRNA-mediated knockdown of MMSET compromised viability of a myeloma cell line, suggesting a biologic role for the protein in malignant cell growth. Collectively, these data suggest that, by acting directly as a modifier of chromatin as well as through binding of other chromatin-modifying enzymes, MMSET influences gene expression and potentially acts as a pathogenic agent in multiple myeloma.
Collapse
|
14
|
Rohner A, Langenkamp U, Siegler U, Kalberer CP, Wodnar-Filipowicz A. Differentiation-promoting drugs up-regulate NKG2D ligand expression and enhance the susceptibility of acute myeloid leukemia cells to natural killer cell-mediated lysis. Leuk Res 2007; 31:1393-402. [PMID: 17391757 DOI: 10.1016/j.leukres.2007.02.020] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 02/19/2007] [Accepted: 02/21/2007] [Indexed: 12/01/2022]
Abstract
Natural killer (NK) cells are potent effectors of innate antitumor defense and are currently exploited for immune-based therapy of human leukemia. However, malignant blood cells in acute myeloid leukemia (AML) display low levels of ligands for the activating immunoreceptor NKG2D and can thus evade NK immunosurveillance. We examined the possibility of up-regulating NKG2D-specific UL16-binding protein (ULBP) ligands using anti-neoplastic compounds with myeloid differentiation potential. Combinations of 5-aza-2'-deoxycytidine, trichostatin A, vitamin D3, bryostatin-1, and all-trans-retinoic acid, used together with myeloid growth factors and interferon-gamma, increased cell surface ULBP expression up to 10-fold in the AML cell line HL60 and in primary AML blasts. Up-regulation of ULBP ligands was associated with induction of myelomonocytic differentiation of AML cells. Higher ULBP expression increased NKG2D-dependent sensitivity of HL60 cells to NK-mediated killing. These findings identify NKG2D ligands as targets of leukemia differentiation therapy and suggest a clinical benefit in combining a pharmacological approach with NK cell-based immunotherapy in AML.
Collapse
Affiliation(s)
- Andreas Rohner
- Department of Research, University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | | | | | | | | |
Collapse
|
15
|
Parekh S, Polo JM, Shaknovich R, Juszczynski P, Lev P, Ranuncolo SM, Yin Y, Klein U, Cattoretti G, Dalla Favera R, Shipp MA, Melnick A. BCL6 programs lymphoma cells for survival and differentiation through distinct biochemical mechanisms. Blood 2007; 110:2067-74. [PMID: 17545502 PMCID: PMC1976344 DOI: 10.1182/blood-2007-01-069575] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The BCL6 transcriptional repressor is the most commonly involved oncogene in diffuse large B-cell lymphomas (DLBCLs). Constitutive expression of BCL6 mediates lymphomagenesis through aberrant proliferation, survival, and differentiation blockade. Binding of BCL6 to the SMRT/N-CoR corepressors mediates the BCL6 survival effect in DLBCL. Although the basis for differentiation blockade is unknown in DLBCL, recent data suggest that BCL6 binding to the MTA3 corepressor might be involved. We report that BCL6 and MTA3 are coexpressed in normal germinal center B cells and DLBCL. Depletion of MTA3 in DLBCL cells induced a differentiation-related BCL6 target gene (PRDM1), but not target genes involved in survival. Accordingly, MTA3 and PRDM1 expression are mutually exclusive in germinal center B cells. We performed chromatin immunoprecipitation (ChIP)-on-chip mapping of the PRDM1 locus, identifying a novel BCL6 binding site on intron 3 of the PRDM1 gene, and show that BCL6 recruits MTA3 to this site. In DLBCL cells, MTA3 depletion induced plasmacytic differentiation but did not decrease viability of DLBCL cells. However, MTA3 depletion synergized with a specific BCL6 inhibitor that blocks SMRT/N-CoR binding to decrease DLBCL viability. Taken together, these results show that BCL6 regulates distinct transcriptional programs through the SMRT/N-CoR and MTA3 corepressors, respectively, and provides a basis for combinatorial therapeutic targeting of BCL6.
Collapse
MESH Headings
- Blotting, Western
- Cell Differentiation
- Cell Survival
- Chromatin Immunoprecipitation
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nuclear Receptor Co-Repressor 1
- Nuclear Receptor Co-Repressor 2
- Oligonucleotide Array Sequence Analysis
- Positive Regulatory Domain I-Binding Factor 1
- Proto-Oncogene Proteins c-bcl-6/genetics
- Proto-Oncogene Proteins c-bcl-6/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA, Small Interfering/pharmacology
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Tissue Array Analysis
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Samir Parekh
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Furchert SE, Lanvers-Kaminsky C, Juürgens H, Jung M, Loidl A, Frühwald MC. Inhibitors of histone deacetylases as potential therapeutic tools for high-risk embryonal tumors of the nervous system of childhood. Int J Cancer 2007; 120:1787-94. [PMID: 17230517 DOI: 10.1002/ijc.22401] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The origin of malignant embryonal tumors is incompletely understood and certain risk groups remain difficult to treat. The epigenetic structure of DNA and its lesions play a role in the origin of these neoplasms. Manipulation of the epigenome may offer novel treatment options. The authors evaluated the cytotoxicity of histone deacetylase inhibitors (HDI) [MS-275, SAHA, TSA, M344, M360, D85, SW55, SW187 and valproic acid (VPA)] on 13 embryonal tumor cell lines [4 medulloblastomas, 5 neuroblastomas, 2 atypical teratoid/rhabdoid tumors (AT/RT), and 2 malignant rhabdoid tumors of the kidney (RTK)] in MTT assay. In addition, HDI effects on hyperacetylation, reexpression of growth regulatory genes and apoptosis were characterized by Western analysis, RT-PCR and annexin-V staining. All HDI inhibited cell proliferation in a time- and dose-dependent manner. VPA was least cytotoxic with GI50 values after 72 hr ranging from 53.6 to 332.9 microM, while TSA was most efficient with GI50 values after 72 hr ranging from 0.01 to 8.8 microM. M344 and M360 were also highly effective. Western blot revealed hyperacetylation of histone H4 after HDI treatment. Reactivation of several genes including the proapoptotic CASP8 was identified by RT-PCR. Annexin-V staining demonstrated a dose and time dependent induction of apoptosis. HDI inhibited the growth of medulloblastoma, neuroblastoma and rhabdoid tumors in vitro. Treatment with HDI induced the reactivation of growth regulatory genes and consequently apoptosis. Our results warrant further studies and may help in the design of new protocols geared at the treatment of high risk embryonal tumors.
Collapse
Affiliation(s)
- Sarah E Furchert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Melnick A. Targeting APL fusion proteins by peptide interference. Curr Top Microbiol Immunol 2007; 313:221-43. [PMID: 17217046 DOI: 10.1007/978-3-540-34594-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A significant barrier to experimental therapeutics is the ability to identify and specifically target oncogenic proteins involved in the molecular pathogenesis of disease. In acute promyelocytic leukemia (APL), aberrant transcription factors and their associated machinery play a central role in mediating the malignant phenotype. The mechanism of action of APL chimeric fusion proteins involves their ability to either self-associate or interact with different partner proteins. Thus, targeting protein-protein interactions could have a significant impact in blocking the activity of APL oncoproteins. As therapeutic targets, the interface between interacting proteins may not always be amenable to highly specific small molecule blockade. In contrast, peptides are well-suited to this purpose and can be reliably delivered when fused to cell-permeable peptide domains. Therapeutic peptides can be designed to directly target APL fusion proteins, their downstream effectors, or other potentially synergistic oncogenic mechanisms of importance in APL blasts. In addition to serving as potential therapeutic agents, such reagents could serve as powerful reagents to dissect the molecular pathogenesis of APL.
Collapse
Affiliation(s)
- A Melnick
- Department of Developmental and Molecular Biology and Medical Oncology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA.
| |
Collapse
|
18
|
Re D, Elter T, Hallek M. Report on the 1st International Workshop of the German Competence Network Malignant Lymphomas. Oncol Res Treat 2007; 30:265-73. [PMID: 17460423 DOI: 10.1159/000100909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The German Competence Network Malignant Lymphomas (KML) is an initiative of the German Federal Ministry of Education and Research with the goal of establishing a cooperative network between lymphoma study groups to support clinical lymphoma research. The network was initiated in 1999 and comprises 9 national lymphoma study groups. More than 8,600 patients are recruited every year into currently 55 clinical trials by more than 2,400 centers in Germany and Europe. It was the specific aim of the 1st international workshop of the KML (March 6 and 7, 2006) in Cologne to bring together international specialists in the field of lymphoma to discuss current concepts of lymphoma pathogenesis and treatment strategies. This report summarizes novel aspects of pathogenesis and targeted approaches, maintenance therapy, management of the elderly patients, and the use of positron emission tomography for clinical decision making.
Collapse
Affiliation(s)
- Daniel Re
- Department of Internal Medicine I, University Hospital of Cologne, Germany
| | | | | |
Collapse
|
19
|
Qian X, LaRochelle WJ, Ara G, Wu F, Petersen KD, Thougaard A, Sehested M, Lichenstein HS, Jeffers M. Activity of PXD101, a histone deacetylase inhibitor, in preclinical ovarian cancer studies. Mol Cancer Ther 2006; 5:2086-95. [PMID: 16928830 DOI: 10.1158/1535-7163.mct-06-0111] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Histone deacetylase inhibitors represent a promising new class of anticancer agents. In the current investigation, we examined the activity of PXD101, a potent histone deacetylase inhibitor, used alone or in combination with clinically relevant chemotherapeutics (docetaxel, paclitaxel, and carboplatin), in preclinical in vitro and in vivo models of ovarian cancer. In vitro activity was examined in ovarian cancer and multidrug-resistant cell lines grown in monolayer culture, and in primary clinical ovarian cancer specimens grown in three-dimensional organoid culture. PXD101 was found to inhibit in vitro cancer cell growth at sub- to low micromolar IC(50) potency, exhibited synergistic activity when used in combination with relevant chemotherapeutics, and effectively inhibited the growth of multidrug-resistant cells. In vivo, PXD101 displayed single-agent antitumor activity on human A2780 ovarian cancer s.c. xenografts which was enhanced via combination therapy with carboplatin. In support of these findings, PXD101 was shown to increase the acetylation of alpha-tubulin induced by docetaxel and the phosphorylation of H2AX induced by carboplatin. Taken together, these results support the clinical evaluation of PXD101 used alone or in combination therapy for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Xiaozhong Qian
- CuraGen Corporation, 322 East Main Street, Branford, CT 06405, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lacayo NJ, DiMartino JF, Wei MC, Dahl GV. CpG Island Methylator Phenotype and Childhood Leukemia. Clin Cancer Res 2006; 12:4787-9. [PMID: 16914562 DOI: 10.1158/1078-0432.ccr-06-0651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Norman J Lacayo
- Department of Pediatrics, Stanford University, Palo Alto, California and Genentech BioOncology, South San Francisco, California, USA
| | | | | | | |
Collapse
|
21
|
|
22
|
Fox BP, Kandpal RP. Transcriptional silencing of EphB6 receptor tyrosine kinase in invasive breast carcinoma cells and detection of methylated promoter by methylation specific PCR. Biochem Biophys Res Commun 2005; 340:268-76. [PMID: 16364251 DOI: 10.1016/j.bbrc.2005.11.174] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Accepted: 11/30/2005] [Indexed: 12/13/2022]
Abstract
The receptor tyrosine kinase EphB6 is expressed at reasonable levels in normal breast cells. It shows decreased abundance in non-invasive breast carcinoma cells and is transcriptionally silenced in invasive breast carcinoma cells. We have characterized EphB6 promoter and correlated the expression of EphB6 transcript to differential methylation of the promoter region. The demethylation of promoter sequence in vivo by growth in media containing 5-aza-2'-deoxycytidine restores the expression of EphB6 to normal levels in breast carcinoma cells, and the ability of the promoter to initiate transcription of a reporter gene is lost after methylation of the promoter sequence. The promoter region has binding sites for various factors such as SP1 and p300. The specific methylation of CpG dinucleotides has allowed us to design primers that can selectively amplify the methylated promoter and thus facilitate identification of normal, non-invasive, and invasive breast cells. The potential significance of EphB6 to serve as a diagnostic and prognostic indicator is discussed.
Collapse
Affiliation(s)
- Brian P Fox
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | | |
Collapse
|
23
|
Mühlisch J, Schwering A, Grotzer M, Vince GH, Roggendorf W, Hagemann C, Sörensen N, Rickert CH, Osada N, Jürgens H, Frühwald MC. Epigenetic repression of RASSF1A but not CASP8 in supratentorial PNET (sPNET) and atypical teratoid/rhabdoid tumors (AT/RT) of childhood. Oncogene 2005; 25:1111-7. [PMID: 16186793 DOI: 10.1038/sj.onc.1209137] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Supratentorial primitive neuroectodermal tumors (sPNET) and atypical teratoid/rhabdoid tumors (AT/RT) of the CNS represent a biological and clinical enigma, despite advances in both molecular techniques and clinical management for these two rare embryonal brain tumors of childhood. Epigenetic changes hold great potential as possible disease mechanisms and may be manipulated therapeutically. We thus studied aberrant methylation of the genes RASSF1A and CASP8 and its consequence on expression in cell lines and primary tumors using a combination of semiquantitative methylation specific PCR (MSP), bisulfite sequencing and RT-PCR. In all, 17 samples of autopsy-derived normal appearing brain served as controls. Opposed to control tissues 19/24 sPNET and 4/6 AT/RT demonstrated aberrant methylation for the RASSF1A promoter region. Treatment of cell lines using 5-Aza-2'-deoxycytidine (5AZA) alone or in combination with trichostatin A (TSA) succeeded in re-establishing expression of RASSF1A in cell lines derived from a renal rhabdoid, an AT/RT and a medulloblastoma. A 5' CpG-rich region of CASP8 was methylated in normal tissues and in tumors. However, CASP8 showed inconsistent expression patterns in normal and tumor tissues. Our results indicate that aberrant methylation of the RASSF1A promoter region may be of importance in the origin and progression of sPNET and AT/RT while the analysed 5'-CpG rich region of the CASP8 gene does not seem to play an important role in these tumors. Further studies of epigenetic changes in these rare tumors are warranted as their biology remains obscure and treatment efforts have been rather unsuccessfull.
Collapse
Affiliation(s)
- J Mühlisch
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Sausville EA, Carducci MA. Making Bad Cells Go Good: The Promise of Epigenetic Therapy. J Clin Oncol 2005; 23:3875-6. [PMID: 15946932 DOI: 10.1200/jco.2005.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|