1
|
Zuo Z. THE1B may have no role in human pregnancy due to ZNF430-mediated silencing. Mob DNA 2023; 14:6. [PMID: 37217947 DOI: 10.1186/s13100-023-00294-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
THE1-family retrovirus invaded the primate genome more than 40 million years ago. Dunn-Fletcher et al. reported one THE1B element upstream of CRH gene alters gestation length by upregulating corticotropin-releasing hormone expression in transgenic mice and concluded it has the same role in human as well. However, no promoter or enhancer mark has been detected around this CRH-proximal element in any human tissue or cell, so probably some anti-viral factor exists in primates to prevents it from wreaking havoc. Here I report two paralogous zinc finger genes, ZNF430 and ZNF100, that emerged during the simian lineage to specifically silence THE1B and THE1A, respectively. Contact residue changes in one finger confers each ZNF the unique ability to preferentially repress one THE1 sub-family over the other. The reported THE1B element contains an intact ZNF430 binding site, thus under the repression of ZNF430 in most tissues including placenta, it is questionable whether or not this retrovirus has any role in human pregnancy. Overall, this analysis highlights the need to study human retroviruses' functions in suitable model system.
Collapse
Affiliation(s)
- Zheng Zuo
- Shenzhen University, Shenzhen, Guangdong, China.
| |
Collapse
|
2
|
Kim JH, Kim WS, Park C. Sildenafil prevents HDACi-induced Epstein-Barr virus reactivation through the PKG pathway in NK/T cell lymphoma; potential implications for HDACi-mediated fatal complications. Antiviral Res 2021; 189:105063. [PMID: 33741394 DOI: 10.1016/j.antiviral.2021.105063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022]
Abstract
Romidepsin, a histone deacetylase (HDAC) inhibitor, has been approved for the treatment of relapsed and refractory peripheral T-cell lymphoma. However the use of romidepsin reportedly causes potent EBV (Epstein-Barr virus) reactivation leading to severe adverse events in patients with natural killer (NK)/T-cell lymphoma (NKTL). As inhibition of EBV lytic cycle reactivation may help prevent romidepsin-induced adverse events in NKTL, we herein set out to identify a safe and effective drug for inhibiting EBV reactivation and examine its mechanism of inhibition. EBV reactivation was evaluated by qRT-PCR of BZLF1 and BRLF1 mRNA expression, qPCR of EBV DNA, and immunoblotting of viral EA-D protein. High-throughput screening of FDA-approved drugs was performed to identify safe and effective molecules and test their effect on romidepsin-induced EBV reactivation in the EBV-positive NKTL cell lines, SNK6 and NK92MI. We found that phosphodiesterase 5 (PDE5) inhibitors, including sildenafil (Viagra; Pfizer), appeared to be nontoxic and effective inhibitors of romidepsin-induced EBV reactivation. Clinical relevance was investigated by qPCR of EBV in two primary effusion samples of NKTL patients. We also investigated the molecular consequences downstream of sildenafil-induced PDE5 inhibition in NKTL cells. A negative correlation was established between the cGMP/PKG pathway and EBV reactivation in NKTL cells. On a molecular level, PDE5 inhibition downregulates BZLF1 and BRLF1 through cGMP/PKG signaling-induced ZNF overexpression. Co-treatment with romidepsin and sildenafil (inhibiting HDAC and PDE5, respectively) showed a synergistic inhibitory effect on NKTL cells, highlighting PDE5 as an attractive target for future therapy in NKTL.
Collapse
Affiliation(s)
- Joo Hyun Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, South Korea
| | - Won Seog Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, South Korea; Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea.
| | - Chaehwa Park
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea.
| |
Collapse
|
3
|
Long-term effect of neonatal inhibition of APP gamma-secretase on hippocampal development in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis 2017; 103:11-23. [PMID: 28359846 PMCID: PMC5439029 DOI: 10.1016/j.nbd.2017.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 03/09/2017] [Accepted: 03/26/2017] [Indexed: 11/21/2022] Open
Abstract
Neurogenesis impairment is considered a major determinant of the intellectual disability that characterizes Down syndrome (DS), a genetic condition caused by triplication of chromosome 21. Previous evidence obtained in the Ts65Dn mouse model of DS showed that the triplicated gene APP (amyloid precursor protein) is critically involved in neurogenesis alterations. In particular, excessive levels of AICD (amyloid precursor protein intracellular domain) resulting from APP cleavage by gamma-secretase increase the transcription of Ptch1, a Sonic Hedgehog (Shh) receptor that keeps the mitogenic Shh pathway repressed. Previous evidence showed that neonatal treatment with ELND006, an inhibitor of gamma-secretase, reinstates the Shh pathway and fully restores neurogenesis in Ts65Dn pups. In the framework of potential therapies for DS, it is extremely important to establish whether the positive effects of early intervention are retained after treatment cessation. Therefore, the goal of the current study was to establish whether early treatment with ELND006 leaves an enduring trace in the brain of Ts65Dn mice. Ts65Dn and euploid pups were treated with ELND006 in the postnatal period P3-P15 and the outcome of treatment was examined at ~ one month after treatment cessation. We found that in treated Ts65Dn mice the pool of proliferating cells in the hippocampal dentate gyrus (DG) and total number of granule neurons were still restored as was the number of pre- and postsynaptic terminals in the stratum lucidum of CA3, the site of termination of the mossy fibers from the DG. Accordingly, patch-clamp recording from field CA3 showed functional normalization of the input to CA3. Unlike in field CA3, the number of pre- and postsynaptic terminals in the DG of treated Ts65Dn mice was no longer fully restored. The finding that many of the positive effects of neonatal treatment were retained after treatment cessation provides proof of principle demonstration of the efficacy of early inhibition of gamma-secretase for the improvement of brain development in DS. Neonatal inhibition of gamma-secretase has long-term effects in a Down syndrome model. Treatment induces long-term restoration of hippocampal neurogenesis and cellularity. Treatment induces long-term restoration of functional connectivity. Treatments with gamma-secretase inhibitors may be exploited for Down syndrome.
Collapse
|
4
|
Jin S, Choi H, Kwon JT, Kim J, Jeong J, Kim J, Hong SH, Cho C. Identification of target genes for spermatogenic cell-specific KRAB transcription factor ZFP819 in a male germ cell line. Cell Biosci 2017; 7:4. [PMID: 28053699 PMCID: PMC5209904 DOI: 10.1186/s13578-016-0132-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/21/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Zfp819, a member of the Krüppel-associated box (KRAB) family, encodes a spermatogenic cell-specific transcription factor. Zfp819-overexpression induces apoptosis and inhibits proliferation in somatic cell lines. RESULTS In the present study, we examined the cellular effects of Zfp819 in a male germ cell line (GC-2 cells). Overexpression of Zfp819 demonstrated an increase in the number of apoptotic cells, leading to inhibition of proliferation in GC-2 cells. We further investigated genes regulated by ZFP819 using microarray analysis and chromatin-immunoprecipitation combined with microarray analysis (ChIP-chip) in GC-2 cells. We identified 118 downregulated genes in Zfp819-overexpressing GC-2 cells using microarray analysis. ChIP-chip assay revealed that 1011 promoter sites (corresponding to 262 genes) were specifically enriched in GC-2 cells transfected with Zfp819. Two genes (trinucleotide repeat containing 6b and annexin A11) were commonly found when we compared the data between microarray and ChIP-chip analyses. Consistent with these results, Zfp819 overexpression significantly reduced the transcript levels of the two genes by binding to their promoter regions. Tissue distribution analysis indicated that both genes were predominantly expressed in testis. It has been reported that these two genes function in apoptosis. CONCLUSION Collectively, our study provides inclusive information on germ cell-specific gene regulation by ZFP819, which is involved in apoptosis, to maintain the integrity of spermatogenesis.
Collapse
Affiliation(s)
- Sora Jin
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| | - Heejin Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| | - Jun Tae Kwon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| | - Jihye Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| | - Juri Jeong
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| | - Jaehwan Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| | - Seong Hyeon Hong
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| | - Chunghee Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005 South Korea
| |
Collapse
|
5
|
Hornick NI, Doron B, Abdelhamed S, Huan J, Harrington CA, Shen R, Cambronne XA, Chakkaramakkil Verghese S, Kurre P. AML suppresses hematopoiesis by releasing exosomes that contain microRNAs targeting c-MYB. Sci Signal 2016; 9:ra88. [PMID: 27601730 DOI: 10.1126/scisignal.aaf2797] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Exosomes are paracrine regulators of the tumor microenvironment and contain complex cargo. We previously reported that exosomes released from acute myeloid leukemia (AML) cells can suppress residual hematopoietic stem and progenitor cell (HSPC) function indirectly through stromal reprogramming of niche retention factors. We found that the systemic loss of hematopoietic function is also in part a consequence of AML exosome-directed microRNA (miRNA) trafficking to HSPCs. Exosomes isolated from cultured AML or the plasma from mice bearing AML xenografts exhibited enrichment of miR-150 and miR-155. HSPCs cocultured with either of these exosomes exhibited impaired clonogenicity, through the miR-150- and miR-155-mediated suppression of the translation of transcripts encoding c-MYB, a transcription factor involved in HSPC differentiation and proliferation. To discover additional miRNA targets, we captured miR-155 and its target transcripts by coimmunoprecipitation with an attenuated RNA-induced silencing complex (RISC)-trap, followed by high-throughput sequencing. This approach identified known and previously unknown miR-155 target transcripts. Integration of the miR-155 targets with information from the protein interaction database STRING revealed proteins indirectly affected by AML exosome-derived miRNA. Our findings indicate a direct effect of AML exosomes on HSPCs that, through a stroma-independent mechanism, compromises hematopoiesis. Furthermore, combining miRNA target data with protein-protein interaction data may be a broadly applicable strategy to define the effects of exosome-mediated trafficking of regulatory molecules within the tumor microenvironment.
Collapse
Affiliation(s)
- Noah I Hornick
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA. Pediatric Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA. Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ben Doron
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA. Pediatric Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA. Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sherif Abdelhamed
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA. Pediatric Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA. Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jianya Huan
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA. Pediatric Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA. Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christina A Harrington
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA. Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rongkun Shen
- Department of Biology, State University of New York, Brockport, NY 14420, USA. Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA. The College at Brockport, State University of New York, Brockport, NY 14420, USA
| | - Xiaolu A Cambronne
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Santhosh Chakkaramakkil Verghese
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA. Pediatric Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA. Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Peter Kurre
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA. Pediatric Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA. Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA. Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
6
|
Ecco G, Cassano M, Kauzlaric A, Duc J, Coluccio A, Offner S, Imbeault M, Rowe HM, Turelli P, Trono D. Transposable Elements and Their KRAB-ZFP Controllers Regulate Gene Expression in Adult Tissues. Dev Cell 2016; 36:611-23. [PMID: 27003935 DOI: 10.1016/j.devcel.2016.02.024] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 02/10/2016] [Accepted: 02/24/2016] [Indexed: 12/21/2022]
Abstract
KRAB-containing zinc finger proteins (KRAB-ZFPs) are early embryonic controllers of transposable elements (TEs), which they repress with their cofactor KAP1 through histone and DNA methylation, a process thought to result in irreversible silencing. Using a target-centered functional screen, we matched murine TEs with their cognate KRAB-ZFP. We found the paralogs ZFP932 and Gm15446 to bind overlapping but distinguishable subsets of ERVK (endogenous retrovirus K), repress these elements in embryonic stem cells, and regulate secondarily the expression of neighboring genes. Most importantly, we uncovered that these KRAB-ZFPs and KAP1 control TEs in adult tissues, in cell culture and in vivo, where they partner up to modulate cellular genes. Therefore, TEs and KRAB-ZFPs establish transcriptional networks that likely regulate not only development but also many physiological events. Given the high degree of species specificity of TEs and KRAB-ZFPs, these results have important implications for understanding the biology of higher vertebrates, including humans.
Collapse
Affiliation(s)
- Gabriela Ecco
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Marco Cassano
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Annamaria Kauzlaric
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Julien Duc
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Andrea Coluccio
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Sandra Offner
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Michaël Imbeault
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Helen M Rowe
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Priscilla Turelli
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland
| | - Didier Trono
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, 1015 Lausanne, Switzerland.
| |
Collapse
|
7
|
Salati S, Zini R, Nuzzo S, Guglielmelli P, Pennucci V, Prudente Z, Ruberti S, Rontauroli S, Norfo R, Bianchi E, Bogani C, Rotunno G, Fanelli T, Mannarelli C, Rosti V, Salmoiraghi S, Pietra D, Ferrari S, Barosi G, Rambaldi A, Cazzola M, Bicciato S, Tagliafico E, Vannucchi AM, Manfredini R. Integrative analysis of copy number and gene expression data suggests novel pathogenetic mechanisms in primary myelofibrosis. Int J Cancer 2016; 138:1657-69. [PMID: 26547506 DOI: 10.1002/ijc.29920] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 10/15/2015] [Accepted: 10/23/2015] [Indexed: 12/27/2022]
Abstract
Primary myelofibrosis (PMF) is a Myeloproliferative Neoplasm (MPN) characterized by megakaryocyte hyperplasia, progressive bone marrow fibrosis, extramedullary hematopoiesis and transformation to Acute Myeloid Leukemia (AML). A number of phenotypic driver (JAK2, CALR, MPL) and additional subclonal mutations have been described in PMF, pointing to a complex genomic landscape. To discover novel genomic lesions that can contribute to disease phenotype and/or development, gene expression and copy number signals were integrated and several genomic abnormalities leading to a concordant alteration in gene expression levels were identified. In particular, copy number gain in the polyamine oxidase (PAOX) gene locus was accompanied by a coordinated transcriptional up-regulation in PMF patients. PAOX inhibition resulted in rapid cell death of PMF progenitor cells, while sparing normal cells, suggesting that PAOX inhibition could represent a therapeutic strategy to selectively target PMF cells without affecting normal hematopoietic cells' survival. Moreover, copy number loss in the chromatin modifier HMGXB4 gene correlates with a concomitant transcriptional down-regulation in PMF patients. Interestingly, silencing of HMGXB4 induces megakaryocyte differentiation, while inhibiting erythroid development, in human hematopoietic stem/progenitor cells. These results highlight a previously un-reported, yet potentially interesting role of HMGXB4 in the hematopoietic system and suggest that genomic and transcriptional imbalances of HMGXB4 could contribute to the aberrant expansion of the megakaryocytic lineage that characterizes PMF patients.
Collapse
Affiliation(s)
- Simona Salati
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Roberta Zini
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Simona Nuzzo
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Valentina Pennucci
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Zelia Prudente
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Samantha Ruberti
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Sebastiano Rontauroli
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Ruggiero Norfo
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Elisa Bianchi
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Costanza Bogani
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Giada Rotunno
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Tiziana Fanelli
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Carmela Mannarelli
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Vittorio Rosti
- IRCCS Policlinico S.Matteo Foundation, Center for the Study of Myelofibrosis, Pavia, Italy
| | | | - Daniela Pietra
- Department of Hematology Oncology, IRCCS Policlinico San Matteo Foundation & University of Pavia, Pavia, Italy
| | - Sergio Ferrari
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Giovanni Barosi
- IRCCS Policlinico S.Matteo Foundation, Center for the Study of Myelofibrosis, Pavia, Italy
| | | | - Mario Cazzola
- Department of Hematology Oncology, IRCCS Policlinico San Matteo Foundation & University of Pavia, Pavia, Italy
| | - Silvio Bicciato
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Rossella Manfredini
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| |
Collapse
|
8
|
Kesh K, Subramanian L, Ghosh N, Gupta V, Gupta A, Bhattacharya S, Mahapatra NR, Swarnakar S. Association of MMP7 -181A→G Promoter Polymorphism with Gastric Cancer Risk: INFLUENCE OF NICOTINE IN DIFFERENTIAL ALLELE-SPECIFIC TRANSCRIPTION VIA INCREASED PHOSPHORYLATION OF cAMP-RESPONSE ELEMENT-BINDING PROTEIN (CREB). J Biol Chem 2015; 290:14391-406. [PMID: 25847246 DOI: 10.1074/jbc.m114.630129] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Indexed: 01/27/2023] Open
Abstract
Elevated expression of matrix metalloproteinase7 (MMP7) has been demonstrated to play a pivotal role in cancer invasion. The -181A→G (rs11568818) polymorphism in the MMP7 promoter modulates gene expression and possibly affects cancer progression. Here, we evaluated the impact of -181A→G polymorphism on MMP7 promoter activity and its association with gastric cancer risk in eastern Indian case-control cohorts (n = 520). The GG genotype as compared with the AA genotype was predisposed (p = 0.02; odds ratio = 1.9, 95% confidence interval = 1.1-3.3) to gastric cancer risk. Stratification analysis showed that tobacco addiction enhanced gastric cancer risk in GG subjects when compared with AA subjects (p = 0.03, odds ratio = 2.46, and 95% confidence interval = 1.07-5.68). Meta-analysis revealed that tobacco enhanced the risk for cancer more markedly in AG and GG carriers. Activity and expression of MMP7 were significantly higher in GG than in AA carriers. In support, MMP7 promoter-reporter assays showed greater transcriptional activity toward A to G transition under basal/nicotine-induced/cAMP-response element-binding protein (CREB) overexpressed conditions in gastric adenocarcinoma cells. Moreover, nicotine (a major component of tobacco) treatment significantly up-regulated MMP7 expression due to enhanced CREB phosphorylation followed by its nuclear translocation in gastric adenocarcinoma cells. Furthermore, chromatin immunoprecipitation experiments revealed higher binding of phosphorylated CREB with the -181G than the -181A allele. Altogether, specific binding of phosphorylated CREB to the G allele-carrying promoter enhances MMP7 gene expression that is further augmented by nicotine due to increased CREB phosphorylation and thereby increases the risk for gastric cancer.
Collapse
Affiliation(s)
- Kousik Kesh
- From the Drug Development Diagnostic and Biotechnology Division, Indian Institute of Chemical Biology, Kolkata 700032
| | - Lakshmi Subramanian
- the Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, and
| | - Nillu Ghosh
- From the Drug Development Diagnostic and Biotechnology Division, Indian Institute of Chemical Biology, Kolkata 700032
| | - Vinayak Gupta
- the Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, and
| | - Arnab Gupta
- the Saroj Gupta Cancer Center and Research Institute, Kolkata 700104, India
| | - Samir Bhattacharya
- the Saroj Gupta Cancer Center and Research Institute, Kolkata 700104, India
| | - Nitish R Mahapatra
- the Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, and
| | - Snehasikta Swarnakar
- From the Drug Development Diagnostic and Biotechnology Division, Indian Institute of Chemical Biology, Kolkata 700032,
| |
Collapse
|
9
|
Prediction of spontaneous regression of cervical intraepithelial neoplasia lesions grades 2 and 3 by proteomic analysis. INTERNATIONAL JOURNAL OF PROTEOMICS 2014; 2014:129064. [PMID: 25018881 PMCID: PMC4082862 DOI: 10.1155/2014/129064] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/29/2014] [Accepted: 05/14/2014] [Indexed: 02/07/2023]
Abstract
Regression of cervical intraepithelial neoplasia (CIN) 2-3 to CIN 1 or less is associated with immune response as demonstrated by immunohistochemistry in formaldehyde-fixed paraffin-embedded (FFPE) biopsies. Proteomic analysis of water-soluble proteins in supernatants of biopsy samples with LC-MS (LTQ-Orbitrap) was used to identify proteins predictive of CIN2-3 lesions regression. CIN2-3 in the biopsies and persistence (CIN2-3) or regression (≤CIN1) in follow-up cone biopsies was validated histologically by two experienced pathologists. In a learning set of 20 CIN2-3 (10 regressions and 10 persistence cases), supernatants were depleted of seven high abundance proteins prior to unidimensional LC-MS/MS protein analysis. Mean protein concentration was 0.81 mg/mL (range: 0.55–1.14). Multivariate statistical methods were used to identify proteins that were able to discriminate between regressive and persistent CIN2-3. The findings were validated in an independent test set of 20 CIN2-3 (10 regressions and 10 persistence cases). Multistep identification criteria identified 165 proteins. In the learning set, zinc finger protein 441 and phospholipase D6 independently discriminated between regressive and persistent CIN2-3 lesions and correctly classified all 20 patients. Nine regression and all persistence cases were correctly classified in the validation set. Zinc finger protein 441 and phospholipase D6 in supernatant samples detected by LTQ-Orbitrap can predict regression of CIN2-3.
Collapse
|
10
|
Coordinated Transcriptional Regulation of Hspa1a Gene by Multiple Transcription Factors: Crucial Roles for HSF-1, NF-Y, NF-κB, and CREB. J Mol Biol 2014; 426:116-35. [DOI: 10.1016/j.jmb.2013.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/09/2013] [Accepted: 09/09/2013] [Indexed: 11/20/2022]
|
11
|
Oliver CH, Nichols J, Watson CJ. The KRAB domain zinc finger protein, Zfp157, is expressed in multiple tissues during mouse embryogenesis and in specific cells in adult mammary gland and skin. Genesis 2013; 51:179-86. [PMID: 23315963 DOI: 10.1002/dvg.22367] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/23/2012] [Accepted: 01/03/2013] [Indexed: 11/07/2022]
Abstract
The functions of members of the large family of transcriptional repressors, the KRAB domain zinc finger proteins, are not well described. We have identified a new member of this family, Zfp157, as a downstream target of the transcription factor Stat6 in mammary gland. Using a gene-trap approach, we have generated mice harboring a Zfp157-LacZ reporter gene. We have characterized the expression of this reporter during mouse embryogenesis and show that it is expressed in the epiblast and subsequently in a number of embryonic tissues including brain, ovary, intestine, kidney, lung, mammary gland, and hair follicle. In the adult, Zfp157 continues to be expressed in a wide range of tissues while specific patterns of reporter gene expression are apparent in the mammary gland, primarily in the basal epithelial cells of ducts and in the sebaceous glands of hair follicles. These data lay the foundation for further work on the function of Zfp157.
Collapse
Affiliation(s)
- Carrie H Oliver
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom
| | | | | |
Collapse
|
12
|
Oliver CH, Khaled WT, Frend H, Nichols J, Watson CJ. The Stat6-regulated KRAB domain zinc finger protein Zfp157 regulates the balance of lineages in mammary glands and compensates for loss of Gata-3. Genes Dev 2012; 26:1086-97. [PMID: 22588720 DOI: 10.1101/gad.184051.111] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Lineage commitment studies in mammary glands have focused on identifying cell populations that display stem or progenitor properties. However, the mechanisms that control cell fate have been incompletely explored. Herein we show that zinc finger protein 157 (Zfp157) is required to establish the balance between luminal alveolar pStat5- and Gata-3-expressing cells in the murine mammary gland. Using mice in which the zfp157 gene was disrupted, we found that alveologenesis was accelerated concomitantly with a dramatic skewing of the proportion of pStat5-expressing cells relative to Gata-3⁺ cells. This suppression of the Gata-3⁺ lineage was associated with increased expression of the inhibitor of helix-loop-helix protein Id2. Surprisingly, Gata-3 becomes dispensable in the absence of Zfp157, as mice deficient for both Zfp157 and Gata-3 lactate normally, although the glands display a mild epithelial dysplasia. These data suggest that the luminal alveolar compartment of the mammary gland is comprised of a number of distinct cell populations that, although interdependant, exhibit considerable cell fate plasticity.
Collapse
Affiliation(s)
- Carrie H Oliver
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | | | | | | | | |
Collapse
|
13
|
GWAS reveals new recessive loci associated with non-syndromic facial clefting. Eur J Med Genet 2012; 55:510-4. [PMID: 22750566 DOI: 10.1016/j.ejmg.2012.06.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 06/09/2012] [Indexed: 01/31/2023]
Abstract
We have applied a GWAS to 40 consanguineous families segregating cases of non-syndromic cleft lip with or without cleft palate (NS CL/P) (a total of 160 affected and unaffected individuals) in order to trace potential recessive loci that confer susceptibility to this common facial malformation. Pedigree-based association test (PBAT) analyses reported nominal evidence of association and linkage over SNP markers located at 11q25 (rs4937877, P = 2.7 × 10(-6)), 19p12 (rs4324267, P = 1.6 × 10(-5)), 5q14.1 (rs4588572, P-value = 3.36 × 10(-5)), and 15q21.1 (rs4774497, P = 1.08 × 10(-4)). Using the Versatile Gene-Based Association Study to complement the PBAT results, we found clusters of markers located at chromosomes 19p12, 11q25, and 8p23.2 overcome the threshold for GWAS significance (P < 1 × 10(-7)). From this study, new recessive loci implicated in NS CL/P include: B3GAT1, GLB1L2, ZNF431, ZNF714, and CSMD1, even though the functional association with the genesis of NS CL/P remains to be elucidated. These results emphasize the importance of using homogeneous populations, phenotypes, and family structures for GWAS combined with gene-based association analyses, and should encourage. other researchers to evaluate these genes on independent patient samples affected by NS CL/P.
Collapse
|
14
|
Huang GJ, He Z, Ma L. ZFP932 suppresses cellular Hedgehog response and Patched1 transcription. VITAMINS AND HORMONES 2012; 88:309-32. [PMID: 22391310 DOI: 10.1016/b978-0-12-394622-5.00014-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Substantial amount of research has been devoted to the understanding of hedgehog (Hh) signal transduction in the past two decades. Although significant progresses have been made on the understanding of individual signaling components of this pathway, the transcriptional regulation of these components is not as well understood. We have recently characterized a novel Krüppel-associated box (KRAB) domain containing zinc finger transcription factor ZFP932 (formerly known as ZNF431) and showed that it transcriptionally regulates several components of the Hh pathway. ZFP932 is part of the largest KRAB-domain zinc finger transcription factor family in the mammalian genome. However, the in vivo functions for the majority of these transcription factors as well as their downstream targets are not fully understood. ZFP932 is a nuclear KRAB-domain zinc finger transcriptional repressor that recruits HDACs to target promoters. Using the embryonic limb mesenchymal cell line MPLB, we uncovered many potential targets for ZFP932. Among them, Patched1 (Ptch1) was found to be a direct transcriptional target of ZFP932. Promoter analyses using reporter assays as well as electrophoretic mobility shift assays revealed three ZFP932 binding sites in the Ptch1 proximal promoter which are bound by ZFP932 in vivo as demonstrated by chromatin immunoprecipitation. Consistently, overexpression of ZFP932 either in MPLB cells or in Xenopus and mouse embryos strongly repressed Ptch1 expression. Conversely, shRNA knockdown of Zfp932 in MPLB cells elevated Ptch1 expression. In addition to regulating Ptch1 transcription, we demonstrated that ZFP932 also regulates hedgehog signaling as Hh pathway readouts are reduced in ZFP932 overexpression and elevated in ZFP932 knockdown MPLB cells. Moreover, perturbation of ZFP932 expression in C3H10T1/2 cells affected Hh-induced osteoblast differentiation. Together, these results implicate ZFP932 in Hh signaling by transcriptionally regulating Hh signal component expression.
Collapse
Affiliation(s)
- G Jason Huang
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | |
Collapse
|