1
|
Tuerxuntayi A, Abulikemu T, Niu C. Mechanisms of 4-Dimethylamino-4'-Methoxy Chalcone in Promoting Melanin Synthesis. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221086895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background and aims: Vitiligo, a pigmentation-associated disease, affects 1–2% of the global population andis difficult to treat. The pathogenetic mechanism of vitiligo remains unclear. Vernonia anthelmintica (L.) Willd. extract for vitiligo treatment was initially recorded 300 years ago. Itschalcone compounds are believed to play essential roles in this treatment. In a previous study, chalcones were shown to enhance melanin production and tyrosinase activity inmouse B16 cells. Materials and methods: In this study, the effects were investigated of 4-dimethylamino-4'-methoxy chalcone (DMC) on theexpression of tyrosinase (TYR), tyrosinase-related protein (TRP)-1, tyrosinase-relatedprotein (TRP)-2, and microphthalmia-associated transcription factor (MITF)on murine B16 cells. Moreover, the signaling pathways of melanogenesis regulation,and the effects of DMCon the AC/cAMP/PKA/CREB (CREB and p-CREB), p38mitogen-activated protein kinase (MAPK) (MAPK, p-p38 MAPK, ERK andp-ERK), Wnt/β-catenin (β-catenin), and SWI/SNF pathways (SOX-10) proteinexpression levels were examined by Western blot. Results: The data showed that DMCcould promote melanin production by upregulating the p-CREB, p-p38,p-ERK and β-catenin proteins.
Collapse
Affiliation(s)
| | | | - Chao Niu
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, China
- University of Chinese Academy of Sciences, Beijing, China
- Nantong Chanyoo Pharmatech Co., Ltd, Nantong, China
| |
Collapse
|
2
|
Wang D, Zhang S, Liu B. TAF5L functions as transcriptional coactivator of MITF involved in the immune response of the clam Meretrix petechialis. FISH & SHELLFISH IMMUNOLOGY 2020; 98:1017-1023. [PMID: 31743760 DOI: 10.1016/j.fsi.2019.11.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 06/10/2023]
Abstract
TAF5L is a component of the P300/CBP-associated factor (PCAF) histone acetylase complex, which serves as a coactivator and takes part in basal transcription such as promoter recognition, complex assembly and transcription initiation. In our study, the full-length sequence of MpTAF5L was identified and characterized in the clam M. petechialis. Sequence analysis showed that the predicted MpTAF5L protein had a N-terminal TAF5-NTD2 domain and a C-terminal WD40-repeats domain. The annotation and evolutionary analysis revealed MpTAF5L had close evolutionary relationship with other invertebrate species. Tissue distribution analysis of TAF5L claimed that it was highly expressed in the mantle, adductor muscle, foot and hepatopancreas. The mRNA expression of MpTAF5L was significantly up-regulated after Vibrio parahaemolyticus challenge, indicating its involvement in the immune response of clam. Yeast two-hybrid assays verified that MpTAF5L can interact with MpMITF (a critical immune-related transcription factor), and our further research clarified this interaction depended upon the N-terminal TAF5-NTD2 domain of MpTAF5L. Moreover, the mRNA expression of MpBcl-2 (a target gene of MITF) was significantly decreased but the mRNA expression of MpMITF was not significantly changed after knockdown of MpTAF5L, which indicated the reduction of MpMITF regulating activity at the same time. These results revealed that MpTAF5L interacted with MpMITF and enhanced the activation of MpMITF, which plays roles in the immune defense against V. parahaemolyticus.
Collapse
Affiliation(s)
- Di Wang
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shujing Zhang
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Baozhong Liu
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
3
|
Role of p300 in the pathogenesis of Henoch-Schonlein purpura nephritis and as a new target of glucocorticoid therapy in mice. Chin Med J (Engl) 2020; 132:1942-1950. [PMID: 31365430 PMCID: PMC6708694 DOI: 10.1097/cm9.0000000000000380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background: Henoch-Schonlein purpura nephritis (HSPN) is a very common secondary kidney disease of childhood. Its pathogenesis and the treatment mechanism of glucocorticoid have not been fully elucidated. The aim of this study was to determine the relationship between p300 and the pathogenesis, glucocorticoid therapy in mice with HSPN, respectively. Methods: Forty-eight C57BL/6N male mice, weighing 18 to 20 g, were selected (3–4 weeks old, n = 8 per group). The mice in the normal control group (Group I) were given normal solvent and the HSPN model group (Group II) were given sensitizing drugs. The mice in Group III were injected intraperitoneally with dexamethasone after being given sensitizing drugs. Meanwhile, mice in Groups IV, V and VI with conditional knockout of p300 were also given normal solvent, sensitizing drugs and dexamethasone. The levels of serum IgA, creatinine, and circulating immune complex (CIC) concentrations, 24 h urinary protein and urinary erythrocyte in C57 wild mice, and p300 conditional knockout mice in each group were measured. The expression of p300 in renal tissues and the expression of glucocorticoid receptor (GR) α and β, transforming growth factor (TGF)-β1, and activator protein (AP)-1 after dexamethasone treatment were determined by real-time polymerase chain reaction and Western blotting. Results: Compared with the normal solvent control group (Group I), the expression of p300 mRNA in the model group (Group II) was significantly up-regulated. Western blotting further confirmed the result. Urinary erythrocyte count, 24 h urinary protein quantification, serum IgA, CIC, and renal pathologic score in Group V were distinctly decreased compared with non-knockout mice in Group II (9.7 ± 3.8 per high-power field [/HP] vs. 18.7 ± 6.2/HP, t = 1.828, P = 0.043; 0.18 ± 0.06 g/24 h vs. 0.36 ± 0.08 g/24 h, t = 1.837, P = 0.042; 18.78 ± 0.85 mg/mL vs. 38.46 ± 0.46 mg/mL, t = 1.925, P = 0.038; 0.80 ± 0.27 μg/mL vs. 1.64 ± 0.47 μg/mL, t = 1.892, P = 0.041; 7.0 ± 0.5 vs. 18.0 ± 0.5, t = 1.908, P = 0.039). Compared with non-knockout mice (Group III), the level of urinary erythrocyte count and serum IgA in knockout mice (Group VI) increased significantly after treatment with dexamethasone (3.7 ± 0.6/HP vs. 9.2 ± 3.5/HP, t = 2.186, P = 0.024; 12.38 ± 0.26 mg/mL vs. 27.85 ± 0.65 mg/mL, t = 1.852, P = 0.041). The expression level of GRα was considerably increased in the knockout group after dexamethasone treatment compared with non-knockout mice in mRNA and protein level (t = 2.085, P = 0.026; t = 1.928, P = 0.035), but there was no statistically significant difference in the expression level of GRβ between condition knockout and non-knockout mice (t = 0.059, P = 0.087; t = 0.038, P = 1.12). Furthermore, the expression levels of glucocorticoid resistance genes (AP-1 and TGF-β1) were notably increased after p300 knockout compared with non-knockout mice in mRNA and protein level (TGF-β1: t = 1.945, P = 0.034; t = 1.902, P = 0.039; AP-1: t = 1.914, P = 0.038; t = 1.802, P = 0.041). Conclusions: p300 plays a crucial role in the pathogenesis of HSPN. p300 can down-regulate the expression of resistance genes (AP-1 and TGF-β1) by binding with GRα to prevent further renal injury and glucocorticoid resistance. Therefore, p300 is a promising new target in glucocorticoid therapy in HSPN.
Collapse
|
4
|
Raja DA, Gotherwal V, Burse SA, Subramaniam YJ, Sultan F, Vats A, Gautam H, Sharma B, Sharma S, Singh A, Sivasubbu S, Gokhale RS, Natarajan VT. pH-controlled histone acetylation amplifies melanocyte differentiation downstream of MITF. EMBO Rep 2020; 21:e48333. [PMID: 31709752 PMCID: PMC6945066 DOI: 10.15252/embr.201948333] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/04/2019] [Accepted: 10/15/2019] [Indexed: 12/20/2022] Open
Abstract
Tanning response and melanocyte differentiation are mediated by the central transcription factor MITF. This involves the rapid and selective induction of melanocyte maturation genes, while concomitantly the expression of other effector genes is maintained. In this study, using cell-based and zebrafish model systems, we report on a pH-mediated feed-forward mechanism of epigenetic regulation that enables selective amplification of the melanocyte maturation program. We demonstrate that MITF activation directly elevates the expression of the enzyme carbonic anhydrase 14 (CA14). Nuclear localization of CA14 leads to an increase of the intracellular pH, resulting in the activation of the histone acetyl transferase p300/CBP. In turn, enhanced H3K27 histone acetylation at selected differentiation genes facilitates their amplified expression via MITF. CRISPR-mediated targeted missense mutation of CA14 in zebrafish results in the formation of immature acidic melanocytes with decreased pigmentation, establishing a central role for this mechanism during melanocyte differentiation in vivo. Thus, we describe an epigenetic control system via pH modulation that reinforces cell fate determination by altering chromatin dynamics.
Collapse
Affiliation(s)
- Desingu Ayyappa Raja
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| | - Vishvabandhu Gotherwal
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| | - Shaunak A Burse
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| | - Yogaspoorthi J Subramaniam
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| | - Farina Sultan
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| | - Archana Vats
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
| | - Hemlata Gautam
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
| | - Babita Sharma
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| | - Sachin Sharma
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
- Present address:
National Institute of ImmunologyNew DelhiIndia
| | - Archana Singh
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| | | | - Rajesh S Gokhale
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Present address:
National Institute of ImmunologyNew DelhiIndia
| | - Vivek T Natarajan
- CSIR‐Institute of Genomics and Integrative BiologyNew DelhiIndia
- Academy of Scientific and Innovative ResearchTaramani, Chennai
| |
Collapse
|
5
|
Kim E, Zucconi BE, Wu M, Nocco SE, Meyers DJ, McGee JS, Venkatesh S, Cohen DL, Gonzalez EC, Ryu B, Cole PA, Alani RM. MITF Expression Predicts Therapeutic Vulnerability to p300 Inhibition in Human Melanoma. Cancer Res 2019; 79:2649-2661. [PMID: 30910803 PMCID: PMC6522293 DOI: 10.1158/0008-5472.can-18-2331] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/27/2018] [Accepted: 03/21/2019] [Indexed: 01/17/2023]
Abstract
Histone modifications, largely regulated by histone acetyltransferases (HAT) and histone deacetylases, have been recognized as major regulatory mechanisms governing human diseases, including cancer. Despite significant effort and recent advances, the mechanism by which the HAT and transcriptional coactivator p300 mediates tumorigenesis remains unclear. Here, we use a genetic and chemical approach to identify the microphthalmia-associated transcription factor (MITF) as a critical downstream target of p300 driving human melanoma growth. Direct transcriptional control of MITF by p300-dependent histone acetylation within proximal gene regulatory regions was coupled to cellular proliferation, suggesting a significant growth regulatory axis. Further analysis revealed forkhead box M1 (FOXM1) as a key effector of the p300-MITF axis driving cell growth that is selectively activated in human melanomas. Targeted chemical inhibition of p300 acetyltransferase activity using a potent and selective catalytic p300/CBP inhibitor demonstrated significant growth inhibitory effects in melanoma cells expressing high levels of MITF. Collectively, these data confirm the critical role of the p300-MITF-FOXM1 axis in melanoma and support p300 as a promising novel epigenetic therapeutic target in human melanoma. SIGNIFICANCE: These results show that MITF is a major downstream target of p300 in human melanoma whose expression is predictive of melanoma response to small-molecule inhibition of p300 HAT activity.
Collapse
Affiliation(s)
- Edward Kim
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Beth E. Zucconi
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | - Muzhou Wu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Sarah E. Nocco
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - David J. Meyers
- Department of Pharmacology & Molecular Sciences, Johns Hopkins School of Medicine, 725 N. Wolfe Street, 316 Hunterian Building, Baltimore, Maryland 21205
| | - Jean S. McGee
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Samantha Venkatesh
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Daniel L. Cohen
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Estela C. Gonzalez
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Byungwoo Ryu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Philip A. Cole
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115;,Corresponding Authors: Rhoda M. Alani 617-638-5517, Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118 (Lead Contact), Philip A. Cole 617-525-5208, Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | - Rhoda M. Alani
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118;,Corresponding Authors: Rhoda M. Alani 617-638-5517, Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118 (Lead Contact), Philip A. Cole 617-525-5208, Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
6
|
Vlčková K, Réda J, Ondrušová L, Krayem M, Ghanem G, Vachtenheim J. GLI inhibitor GANT61 kills melanoma cells and acts in synergy with obatoclax. Int J Oncol 2016; 49:953-60. [PMID: 27572939 DOI: 10.3892/ijo.2016.3596] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/26/2016] [Indexed: 11/06/2022] Open
Abstract
MEK kinase inhibitors (trametinib and selumetinib) or kinase inhibitors directed against mutated BRAF(V600E) (vemurafenib and dabrafenib) have initial encouraging effects in the treatment of melanoma but acquired resistance appears almost invariably after some months. Studies revealed mutually exclusive NRAS and BRAF activating mutations driving the MAPK/ERK pathway among human melanomas. Although combination therapy exerts significantly better antitumor cell efficacy, complete remission is rarely achieved. To employ an alternative approach, we have targeted the Hedgehog/GLI pathway, which is deregulated in melanomas, through the GLI1/2 inhibitor GANT61, alone or accompanied with the treatment by the BCL2 family inhibitor obatoclax in 9 melanoma cell lines. Thus, we targeted melanoma cells irrespective of their NRAS or BRAF mutational status. After GANT61 treatment, the cell viability was drastically diminished via apoptosis, as substantial nuclear DNA fragmentation was detected. In all tested melanoma cell lines, the combined treatment was more efficient than the application of each drug alone at the end of the cell growth with inhibitors. GANT61 was efficient also alone in most cell lines without the addition of obatoclax, which had only a limited effect when used as a single drug. In most cell lines, tumor cells were eradicated after 5-9 days of combined treatment in colony outgrowth assay. To conclude, GANT61 treatment might become a hopeful and effective anti-melanoma targeted therapy, especially when combined with the BCL2 family inhibitor obatoclax.
Collapse
Affiliation(s)
- Kateřina Vlčková
- Laboratory of Transcription and Cell Signaling, Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Jiri Réda
- Laboratory of Transcription and Cell Signaling, Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Lubica Ondrušová
- Laboratory of Transcription and Cell Signaling, Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Mohammad Krayem
- LOCE-Institut Jules Bordet, Université Libre de Bruxelles, B-1000 Brussels, Belgium
| | - Ghanem Ghanem
- LOCE-Institut Jules Bordet, Université Libre de Bruxelles, B-1000 Brussels, Belgium
| | - Jiri Vachtenheim
- Laboratory of Transcription and Cell Signaling, Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
7
|
Howlin J, Cirenajwis H, Lettiero B, Staaf J, Lauss M, Saal L, Borg Å, Gruvberger-Saal S, Jönsson G. Loss of CITED1, an MITF regulator, drives a phenotype switch in vitro and can predict clinical outcome in primary melanoma tumours. PeerJ 2015; 3:e788. [PMID: 25755924 PMCID: PMC4349148 DOI: 10.7717/peerj.788] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/04/2015] [Indexed: 12/15/2022] Open
Abstract
CITED1 is a non-DNA binding transcriptional co-regulator whose expression can distinguish the ‘proliferative’ from ‘invasive’ signature in the phenotype-switching model of melanoma. We have found that, in addition to other ‘proliferative’ signature genes, CITED1 expression is repressed by TGFβ while the ‘invasive’ signature genes are upregulated. In agreement, CITED1 positively correlates with MITF expression and can discriminate the MITF-high/pigmentation tumour molecular subtype in a large cohort (120) of melanoma cell lines. Interestingly, CITED1 overexpression significantly suppressed MITF promoter activation, mRNA and protein expression levels while MITF was transiently upregulated following siRNA mediated CITED1 silencing. Conversely, MITF siRNA silencing resulted in CITED1 downregulation indicating a reciprocal relationship. Whole genome expression analysis identified a phenotype shift induced by CITED1 silencing and driven mainly by expression of MITF and a cohort of MITF target genes that were significantly altered. Concomitantly, we found changes in the cell-cycle profile that manifest as transient G1 accumulation, increased expression of CDKN1A and a reduction in cell viability. Additionally, we could predict survival outcome by classifying primary melanoma tumours using our in vitro derived ‘CITED1-silenced’ gene expression signature. We hypothesize that CITED1 acts a regulator of MITF, functioning to maintain MITF levels in a range compatible with tumourigenesis.
Collapse
Affiliation(s)
- Jillian Howlin
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden.,Cell and Experimental Pathology, Department of Laboratory Medicine Malmö, Lund University, Sweden
| | - Helena Cirenajwis
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| | - Barbara Lettiero
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| | - Johan Staaf
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| | - Martin Lauss
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| | - Lao Saal
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| | - Åke Borg
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| | - Sofia Gruvberger-Saal
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| | - Göran Jönsson
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Scheelevägen, Lund, Sweden
| |
Collapse
|
8
|
Mou Y, Jiang X, Du Y, Xue L. Intelligent bioengineering in vitiligo treatment: Transdermal protein transduction of melanocyte-lineage-specific genes. Med Hypotheses 2012; 79:786-9. [DOI: 10.1016/j.mehy.2012.08.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 08/18/2012] [Accepted: 08/25/2012] [Indexed: 11/16/2022]
|
9
|
Vachtenheim J, Borovanský J. “Transcription physiology” of pigment formation in melanocytes: central role of MITF. Exp Dermatol 2010; 19:617-27. [PMID: 20201954 DOI: 10.1111/j.1600-0625.2009.01053.x] [Citation(s) in RCA: 284] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
10
|
Vachtenheim J, Ondrusová L, Borovanský J. SWI/SNF chromatin remodeling complex is critical for the expression of microphthalmia-associated transcription factor in melanoma cells. Biochem Biophys Res Commun 2010; 392:454-9. [PMID: 20083088 DOI: 10.1016/j.bbrc.2010.01.048] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 01/13/2010] [Indexed: 11/28/2022]
Abstract
The microphthalmia-associated transcription factor (MITF) is required for melanocyte development, maintenance of the melanocyte-specific transcription, and survival of melanoma cells. MITF positively regulates expression of more than 25 genes in pigment cells. Recently, it has been demonstrated that expression of several MITF downstream targets requires the SWI/SNF chromatin remodeling complex, which contains one of the two catalytic subunits, Brm or Brg1. Here we show that the expression of MITF itself critically requires active SWI/SNF. In several Brm/Brg1-expressing melanoma cell lines, knockdown of Brg1 severely compromised MITF expression with a concomitant downregulation of MITF targets and decreased cell proliferation. Although Brm was able to substitute for Brg1 in maintaining MITF expression and melanoma cell proliferation, sequential knockdown of both Brm and Brg1 in 501mel cells abolished proliferation. In Brg1-null SK-MEL-5 melanoma cells, depletion of Brm alone was sufficient to abrogate MITF expression and cell proliferation. Chromatin immunoprecipitation confirmed the binding of Brg1 or Brm to the promoter of MITF. Together these results demonstrate the essential role of SWI/SNF for expression of MITF and suggest that SWI/SNF may be a promissing target in melanoma therapy.
Collapse
Affiliation(s)
- Jiri Vachtenheim
- Laboratory of Molecular Biology, University Hospital, Charles University, Prague, Czech Republic.
| | | | | |
Collapse
|
11
|
Sestáková B, Ondrusová L, Vachtenheim J. Cell cycle inhibitor p21/ WAF1/ CIP1 as a cofactor of MITF expression in melanoma cells. Pigment Cell Melanoma Res 2010; 23:238-51. [PMID: 20067556 DOI: 10.1111/j.1755-148x.2010.00670.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
p21/ WAF1/ Cip1 (p21), a cyclin-dependent kinase inhibitor, may act as an antioncogene, but may also behave as a tumor promoting factor by inhibiting apoptosis. p21 is also a transcriptional regulator, exerting this activity independently of cyclin-dependent kinases. Increased p21 protein levels were found in a subset of melanomas. However, the mechanism(s) contributing to the tolerance of high p21 levels in melanoma cells remains unexplained. Here, we show that the p21 protein positively regulates the promoter of microphthalmia-associated transcription factor (MITF), a transcription factor which plays a central role in the expression of melanocyte-specific genes, lineage determination, and survival of melanoma cells. p21 activated the MITF promoter-reporter, occupied the promoter in vivo and cooperated with cAMP response element binding protein (CREB) in promoter activation. In addition, p21 knockdown by shRNA resulted in a decrease of MITF protein and promoter activity, and p21 protein levels correlated with MITF mRNA in most cell lines tested. As the p21 gene is a known transcriptional target of MITF, the reciprocal stimulation of transcription may constitute a positive-feedback loop reinforcing MITF expression in melanoma cells. Our results might help explain the tolerance of increased p21 levels found in some melanomas.
Collapse
Affiliation(s)
- Blanka Sestáková
- Laboratory of Molecular Biology, University Hospital, Charles University, Prague, Czech Republic
| | | | | |
Collapse
|
12
|
Liu F, Fu Y, Meyskens FL. MiTF regulates cellular response to reactive oxygen species through transcriptional regulation of APE-1/Ref-1. J Invest Dermatol 2008; 129:422-31. [PMID: 18971960 DOI: 10.1038/jid.2008.255] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microphthalmia-associated transcription factor (MiTF) is a key transcription factor for melanocyte lineage survival. Most previous work on this gene has been focused on its role in development. A role in carcinogenesis has emerged recently, but the mechanism is unclear. We classified melanoma cells into MiTF-positive and -negative groups and explored the function of MiTF in regulating cellular responses to reactive oxygen species (ROS). The MiTF-positive melanoma cell lines accumulated high levels of apurinic/apyrimidinic endonuclease (APE-1/Ref-1, redox effector-1), a key redox sensor and DNA endonuclease critical for oxidative DNA damage repair. We demonstrate that APE-1 is a transcriptional target for MiTF. Knocking down MiTF led to reduced APE-1 protein accumulation, as well as abolished induction of APE-1 by ROS. MiTF-negative melanoma cells survived more poorly under ROS stress than the MiTF-positive cells based on 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and Trypan blue staining. Overexpression of APE-1 partially rescued ROS-induced cell death when MiTF was depleted. We conclude that MiTF regulates cellular response to ROS by regulation of APE-1, and this may provide a mechanism of how MiTF is involved in melanoma carcinogenesis.
Collapse
Affiliation(s)
- Feng Liu
- Department of Medicine, Chao Family Comprehensive Cancer Center, Irvine School of Medicine, University of California, Orange, California 92868, USA.
| | | | | |
Collapse
|